+ All Categories
Home > Documents > University of Groningen Early onset sepsis in Suriname ...

University of Groningen Early onset sepsis in Suriname ...

Date post: 08-Jan-2022
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
184
University of Groningen Early onset sepsis in Suriname Zonneveld, Rens IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2017 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Zonneveld, R. (2017). Early onset sepsis in Suriname: Epidemiology, Pathophysiology and Novel Diagnostic Concepts. Rijksuniversiteit Groningen. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license. More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne- amendment. Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 08-01-2022
Transcript

University of Groningen

Early onset sepsis in SurinameZonneveld, Rens

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2017

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Zonneveld, R. (2017). Early onset sepsis in Suriname: Epidemiology, Pathophysiology and NovelDiagnostic Concepts. Rijksuniversiteit Groningen.

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license.More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne-amendment.

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 08-01-2022

Early Onset Sepsis in SurinameEpidemiology, Pathophysiology, and Novel Diagnostic Concepts

Rens Zonneveld

ISBN (printed): 978-94-034-0256-7

ISBN (digital): 978-94-034-0257-4

Cover design: Frans Mettes

Lay-out and Printing: Off Page, Amsterdam

© 2017, Rens Zonneveld

All rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means

without permission of the author.

Financial support for the research in this thesis is greatly acknowledged. The following institutes and organisations provided

funding for completion and printing of this thesis:

  

Stichting ‘De Drie Lichten’. 

Early Onset Sepsis in Suriname

Epidemiology, Pathophysiology, and Novel Diagnostic Concepts

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de rector magnificus prof. dr. E. Sterken

en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op

maandag 11 december 2017 om 16.15 uur

door

Rens Zonneveld

geboren op 8 april 1983 te Breda

Early Onset Sepsis in Suriname

Epidemiology, Pathophysiology, and Novel Diagnostic Concepts

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de rector magnificus prof. dr. E. Sterken

en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op

maandag 11 december 2017 om 16.15 uur

door

Rens Zonneveld

geboren op 8 april 1983 te Breda

Promotor

Prof. dr. G. Molema

Copromotores

Dr. F.B. Plötz

Dr. M. van Meurs

Beoordelingscommissie

Prof. dr. J.M. Smit

Prof. dr. J.B. van Woensel

Prof. dr. J.G. Zijlstra

TABLE OF CONTENTSChapter 1 General Introduction and Thesis Outline 7

Part I Epidemiology of Early Onset Sepsis in Suriname 23

Chapter 2 Improved Referral and Survival of Newborns after Scaling Up of

Intensive Care in Suriname 27

BMC Pediatrics, Accepted for Publication

Part II Prediction of Early Onset Sepsis 47

Chapter 3 Association between Early Onset Sepsis Calculator and

Infection Parameters for Newborns with Suspected Early Onset Sepsis 51

J Clin Neonatol 2017, 6:159-62

Chapter 4 Immature-to-total-granulocyte Ratio as a Guide for Antibiotic Treatment in

Suspected Early Onset Sepsis in Surinamese Newborns 59

Submitted

Part III The Vascular Pathophysiology of Early Onset Sepsis 71

Chapter 5 Soluble Adhesion Molecules as Markers for Sepsis and the Potential

Pathophysiological Discrepancy in Neonates, Children and Adults 75

Critical Care 2014, 18:204

Chapter 6 Early Onset Sepsis in Surinamese Newborns is Not Associated with

Elevated Serum Levels of Endothelial Cell Adhesion Molecules and

Their Shedding Enzymes 99

Submitted

Chapter 7 Low Serum Angiopoietin-1, high Angiopoietin-2, and high Ang-2/Ang-1

Protein Ratio are Associated with Early Onset Sepsis in Surinamese Newborns 119

Shock 2017, May 22

Chapter 8 Analyzing Neutrophil Morphology, Mechanics, and Motility in Sepsis:

Options and Challenges for Novel Bedside Technologies 133

Critical Care Medicine 2016, 44:218-28

Chapter 9 Summary & Future Perspectives 157

Appendices 169

Appendix I Letter to the editor Critical Care (Critical Care 2016, 20:235-36.) 171

Appendix II Samenvatting (Summary in Dutch) 174

Appendix III Dankwoord 176

Appendix IV List of Publications 180

Appendix V Curriculum Vitae 182

Promotor

Prof. dr. G. Molema

Copromotores

Dr. F.B. Plötz

Dr. M. van Meurs

Beoordelingscommissie

Prof. dr. J.M. Smit

Prof. dr. J.B. van Woensel

Prof. dr. J.G. Zijlstra

1 General Introduction and Thesis Outline

GEN

ERA

L INTRO

DU

CTIO

N

9

1A COMMON CASE OF SUSPECTED EARLY ONSET SEPSIS IN SURINAMEA day prior to giving birth the mother had taken a boat from her village downstream the Suriname

River to the nearest mission post in Debike1. She had been pregnant for eight full moons. Her water

had broken a few days earlier, but the baby had not arrived yet. The friendly datra2 at the mission

post phoned somebody in the city of Paramaribo and spoke Bakratongo3. People in the village had

talked about the new at’oso4 for babies. Many women went there to give birth and they brought

her there too. After six hours she arrived and spent the night in a room with four other women.

She felt like she had korsu5.

Her daughter was born the next day and although she was crying loudly they still took her to

the baby hospital. Doctors and nurses were standing around a glass box that held her daughter.

The doctors seemed confused. One of the nurses spoke her tongo6 and explained that her baby

was doing fine but could have an infection. They had taken her daughter’s blood to see if it was

infected. Depending on her daughter’s condition and the test results they were going to decide

whether to continue the antibiotics they had started. The nurse said her daughter could suffer

from sepsis, wan takru siki fu brudu7.

In the next few days she spent many hours next to the glass box in the spacious baby room.

To her, her daughter seemed healthy and the same as her four earlier children. After three days,

the doctors used a nanai8 to take her brudu9 for the second time. The nurse told her the results

were fine. However, they were still going to finish her treatment with more antibiotics. Finally,

after a total of seven days they started their long journey home.

In this thesis, I focus on newborns admitted to the only neonatal intensive care unit (NICU) in

Suriname, which is located in Paramaribo, with a specific focus on dilemmas of Early Onset Sepsis –

from epidemiology and prediction towards changes in vascular endothelial integrity, principles of

leukocyte-endothelial interaction, and novel diagnostic methodologies for its timely recognition

or exclusion.

Rens Zonneveld, M.D.

July 2017

1 Village located along the Suriname River in the district Sipaliwini in the interior of Suriname.

Translated from the Surinamese language (Sranan Tongo)2 physician;3 Dutch;4 hospital;5 fever;6 language; 7 a serious infection of the blood;8 needle;9 blood.

GEN

ERA

L INTRO

DU

CTIO

N

10

1EARLY ONSET SEPSIS Early onset sepsis (EOS) is defined as onset of sepsis in newborns within 72 hours after birth [1]. When

intra-uterine infection is present, the fetus can become infected due to increased permeability of

the skin and mucosa for bacterial invasion. EOS is also caused by vertical transmission of pathogens

in the vaginal canal from mother to fetus during labor.

EOS is a leading cause of morbidity and mortality amongst newborns [1-6]. In Western (i.e.,

North American and European) countries incidence of blood culture proven EOS ranges from

0.01 to about 1.2 per 1000 live births. Incidence rates of EOS increase with decreasing gestational

age and birth weight, with the highest incidence (i.e., 26 per 1000 live births) and mortality (i.e.,

50-60% of blood culture proven cases) amongst infants with a birth weight below 1000 grams

(2-4). EOS is associated with colonization of the birth canal (about 30% of mothers in Western

countries) with Group B Streptococcus (GBS) [1]. In Western countries over 45% of all cases of

culture proven EOS GBS (45%) is the responsible pathogen, followed by Escherichia coli (E.coli)

(25%) (5,6). Other bacteria that cause EOS include Listeria Monocytogenes, gram-negative enteric

bacilli (i.e., Enterobacter spp., Klebsiella spp.) and Enterococcus spp. [1]. Viruses (predominantly

entero and herpes simplex virus) are also identified causes of EOS [1].

After the introduction of intrapartum antibiotics as prophylaxis for GBS, incidence of EOS has

decreased about 10-fold over the last 20 years in many Western countries and South Africa [7].

However, recent data indicates that, while incidence of EOS due to GBS is decreasing, incidence

of EOS with E.coli increases, probably due to altered resistance patterns of E.coli strains [1,5,8].

Additionally, GBS prevention approaches may have contributed to the rise of multi resistant gram-

positive strains, such as Methicillin resistant Staphylococcus aureus, as causes for EOS [1,9-11].

Maternal GBS vaccination to further reduce maternal GBS colonization and incidence of EOS, while

preventing antibiotic exposure, is currently under investigation [12].

EARLY ONSET SEPSIS IN THE NON-WESTERN WORLDStudies of EOS in low resource settings in the non-Western world are severely underrepresented

in the literature [13-16]. The vast majority of data on EOS are from upper-middle to high-income

countries in North America and Europe. Despite the lack of detailed data on EOS in the non-

Western world, there is a strong indication that over 90% of global neonatal deaths due to EOS

occurs in these low-to-middle income countries [17,18]. Large meta-analyses revealed incidence

of EOS in low-income countries at least similar to Western countries [13,15,16]. However, in these

analyses low-income countries represented only 5-10% of the total data leaving the true global

impact of EOS underestimated. Additionally, underdiagnosing (i.e., due to lack of resources and

logistic or financial constraints) and underreporting of EOS are common issues in low-resource

settings further enhancing underestimation of the true global impact of EOS [19,20]. Furthermore,

due to limited local availability of proper laboratory facilities, studies from these countries often

lack blood culture confirmed results. As a result, the spectrum of bacterial pathogens involved

in EOS in the non-Western world remains relatively unclear. More data on incidence, causative

organisms, morbidity and mortality from non-Western countries remain critical before proper

GEN

ERA

L INTRO

DU

CTIO

N

11

1prevention strategies and clinical management of suspected EOS can be achieved. Additionally,

since there is strong indication that incidence rates of culture proven EOS are substantially higher

in the non-Western world versus the Western world, studies from non-Western countries may

contribute immensely to our knowledge on basic and pathophysiological principles of EOS.

EARLY ONSET SEPSIS IN SURINAMESuriname is small developing country on the Northeastern corner of South America with

a multiethnic population of about 550,000 people [21]. About half of the population of Suriname

lives in its capital, the city of Paramaribo. Medical care is provided by four hospitals in Paramaribo,

namely the Academic Hospital Paramaribo, ‘s Lands Hospital, Diakonessen Hospital and St.

Vincentius Hospital, and the Streekziekenhuis in Nickerie. Suriname has an annual birth rate

of approximately 10,000 births. Over 90% of these births take place at the maternity wards of

the hospitals in Paramaribo. In rural parts of Suriname Medical Mission Posts provide primary

health care to the inhabitants, including basic obstetric care.

The earliest data on neonatal mortality in Suriname dates back to the detailed documentations

by Dr. Paul Christiaan Flu (1884 (Paramaribo, Suriname) - 1945 (Leiden, The Netherlands)) from

the early 20th century. In his seminal, yet forgotten, work Flu describes the poor socio-economic

circumstances after over three centuries of slavery and its effect on neonatal and pediatric care

and mortality rates [22]. Between 1900 and 1909, 9,259 live births were recorded of whom 474

died within the first 14 days of life, making a high average death rate of 51.2 per 1000 live births

for that age category. Over half (N=284) of these deaths were the result of pre- and dysmaturity,

yet about one third (N=110) of these deaths were from unknown cause and potentially following

neonatal infection.

Currently, neonatal death rate, defined as death within the first month of life, in Suriname has

decreased, but remains high with 12.9 per 1000 live births [23]. Early neonatal death (i.e., death

within the first 7 days of life) is estimated at 16 per 1000 live births [24]. Preliminary data from

the Suriname Perinatal and Infant Mortality Survey estimates contribution of infection to early

neonatal mortality at 24% (4 per 1000 live births) of all early neonatal deaths [23]. In contrast, in

The Netherlands incidence of EOS alone was 0.19 per 1000 live births in 2014 [25].

These numbers indicate a high burden of neonatal infection in Suriname. About 40 newborns

die each year of infection. Despite the overall idea of the impact of infectious disease in Surinamese

newborns, detailed information regarding incidence, type of infection (i.e., EOS versus LOS),

microbial causes, mortality and morbidity, antibiotic treatment (type and duration), and exact

epidemiological determinants are currently unavailable. In Chapter 2 of this thesis we explore

the epidemiology and outcomes of newborns admitted to Suriname’s neonatal care facility at

the Academic Hospital Paramaribo. This facility was established in 2008 and renewed in 2015 with

expansion of intensive care capacity, training of personnel and new equipment. For this chapter

we hypothesized that tertiary function and morbidity and mortality rates of treated newborns

would improve after the transition to the renewed neonatal care facility. Additionally, the impact

of EOS on mortality of Surinamese newborns is explored.

GEN

ERA

L INTRO

DU

CTIO

N

12

1EARLY ONSET SEPSIS: A DIAGNOSTIC AND THERAPEUTIC DILEMMAEOS can present with relatively mild symptoms resulting in late discovery with high risk for

mortality and morbidity. Furthermore, clinical symptoms of EOS are extremely diverse and difficult

to distinguish from physiologic symptoms of neonatal transition from intra-to-extrauterine life

and other non-infectious neonatal disease [3,9,26]. This complicates clinical decision-making on

start and duration of antibiotic treatment leading to significant overtreatment. For example, in

the European Union almost 8% of newborns are treated with antibiotics for suspected EOS, while

incidence rates of bacterial culture proven EOS range from 0.01 to 0.53 per 1000 live births in those

countries [3].

Blood culturing is considered the golden standard diagnostic test for EOS and takes several

days to become positive. Upon suspicion of EOS, newborns are observed and treated empirically

for EOS with antibiotics for at least 48 hours until results of blood culturing are known [1]. However,

blood cultures are only positive in 0.01 to 1.2 per 1000 live births in countries in the European

Union and North America. Contributing to this low prevalence may be false negativity due to low

yield of bacteria in low sample volumes or low-density bacteremia in general. Nonetheless, over

60% of newborns empirically treated with antibiotics for suspected EOS are treated for longer

than 72 hours even when blood cultures are negative [27]. Antibiotic stewardship is necessary to

reduce this overtreatment [28].

These dilemmas in the management of EOS pose a huge cost and socioeconomic threat,

especially in non-Western countries [1,6,16]. Moreover, it is becoming clear that prolonged

treatment of newborns with antibiotics also can negatively and severely impact early and

long-term neurodevelopment, growth, the developing immune system, and gut microbiome

resistance patterns [29-32].

CURRENT APPROACHES IN PREDICTION OF EARLY ONSET SEPSISSince clinical presentation and blood culturing have poor specificity for EOS, additional approaches

to aid clinical decision-making whether to start and/or continue antibiotic treatment have been

developed in the recent decade. Approaches that are commonly used in the clinic include maternal

risk factor stratification and serial measurement of C-reactive protein (CRP) levels and leukocyte

counts. Each of these has limitations in clinical utility, as will be discussed below.

Maternal Risk Factor Stratification

Maternal risk factors for EOS (i.e., presence and duration of prolonged rupture of the membranes,

intrapartum fever or administration of antibiotics, and presence of maternal GBS colonization,

as the most common cause of EOS in Western countries, have been used to predict presence of

EOS in newborns. In an attempt to overcome the problem of antibiotic overtreatment amongst

near and at term newborns with a gestational age equal or above 34 weeks, a risk stratification

strategy based on these factors and neonatal clinical findings has been developed in 2010 by

Escobar et al., which was revised in 2014 (33). This EOS calculator (available online at https://

GEN

ERA

L INTRO

DU

CTIO

N

13

1neonatalsepsiscalculator.kaiserpermanente.org) provides a quantitative estimation of EOS

risk along with a recommendation whether to start antibiotic treatment. Since its inception,

two retrospective studies revealed that application of the EOS calculator might help to reduce

antibiotic therapy with 50% (34,35). Additionally, the EOS calculator uses local incidence rates of

EOS as a variable, which still have to be established in many non-Western countries.

Correlation of results of the EOS calculator with biomarkers of inflammation in the newborn

may be helpful in further increasing its clinical utility. Therefore, the study in Chapter 3 explores

the relationship of results from the EOS calculator with results of serial measurement of CRP and

leukocyte counts in a cohort of Dutch near and at term newborns. For this study we hypothesized

that higher EOS calculator result, indicating higher risk for EOS, corresponds with an increase in

CRP and low leukocyte counts.

C-reactive Protein

CRP is an endogenous acute phase reactant synthesized by the liver upon infection [36]. Serum

CRP in newborns always represents endogenous synthesis since it passes the placenta in extremely

low quantities [37]. CRP is constitutively present in serum of newborns at very low concentrations

and its levels are dependent on gestational age and birth weight. CRP synthesis starts immediately

after an inflammatory stimulus by chemokines, such as interleukin (IL)-1, and IL-6, with serum

concentrations rising above the usual laboratory threshold of 5 mg/L after 6 hours and peaking

after 48 hours. This delayed synthesis results in poor sensitivity of CRP levels during early EOS.

In most practices, in the newborn suspected and treated with antibiotics for EOS, a repeat CRP

level below the laboratory threshold measured between 24 to 48 hours after start of antibiotics

has negative predictive value of 99% for EOS, yet only in case of a negative blood culture plus

a clinically improved newborn [37]. However, in clinical practice, despite this strong negative

value, the repeat CRP also leads to even more testing, culturing, and longer treatment duration

and hospital stay (38).

Leukocyte Counts

Inflammation and infection causes release of leukocytes from the bone marrow into the circulation.

Leukocyte counts (both total and subset, predominantly neutrophil, counts) have been widely

used to assess EOS [1,3]. However, both leukocyte and neutrophil counts lack specificity for

prediction of EOS [39,40]. Their numbers are dependent on many perinatal factors such as

gestational age, birth weight, type of delivery, and post partum age [41]. Neutropenia has shown

the most specificity for EOS [42]. However, as discussed above, due to low prevalence of positive

blood cultures, clinical decision-making on start and duration of antibiotic treatment is often

based on non-specific clinical symptoms and repeated measurement of CRP. Serial measurement

of low immature-to-total granulocyte (I/T) ratio has been showen to have a negative predictive

value for blood culture positive EOS of 99% [42]. Chapter 4 explores the relevance of a one-point

automated I/T ratio determination in prediction of duration of antibiotic therapy in a retrospective

cohort of Surinamese newborns with suspected EOS. For this study, we hypothesized that early

GEN

ERA

L INTRO

DU

CTIO

N

14

1establishment of a one-point low I/T ratio is associated with short duration of antibiotic treatment

in suspected EOS. This may prevent start of unnecessary antibiotic treatment, which may help to

reduce the antibiotic burden in developing countries.

EARLY ONSET SEPSIS: A NEED FOR NOVEL DIAGNOSTIC STRATEGIESThe approaches described above have been used for over 20 years and have remained virtually

unchanged. A recent international survey established that in practice only 31% of clinicians use

CRP levels and leukocyte counts as arguments for the decision to start antibiotics [43]. Many other

biomarkers have been investigated, but have not made it into the clinic for various reasons such as

poor specificity, short half lives of biomarkers, lack of reproducibility, or technical issues [44]. At

this point, serial measurement of procalcitonin, an acute phase reactant similar to CRP, is showing

promise in negative prediction of EOS and reduction of antibiotic treatment in Western countries

[45]. However, novel and practical approaches for early and prompt confirmation or exclusion of

EOS remain necessary to reduce antibiotic overtreatment, while improving outcomes. Elements

of the vascular pathophysiology may be relevant for development of these novel approaches,

which will be discussed below.

THE VASCULAR PATHOPHYSIOLOGY OF EARLY ONSET SEPSISThe diagnostic and therapeutic dilemmas of EOS occur, at least in part, because its pathophysiology

remains poorly understood. Endothelial inflammatory activation and leukocyte-endothelial

interactions are key processes in sepsis pathophysiology. Part 3 is aimed to provide more insight

into these processes in newborns to unravel aspects of EOS pathophysiology and provide novel

concepts for its timely diagnosis and management.

LEUKOCYTE-ENDOTHELIAL INTERACTIONS: SHEDDING OF ADHESION MOLECULES IN EARLY ONSET SEPSISLeukocyte-endothelial interactions are involved in any infectious pathophysiology [46]. A body

of evidence is indicating that aberrant leukocyte, mostly neutrophil, activation and recruitment

towards the endothelium plays a pivotal role in breakdown of the vascular endothelium, which, in

turn, is associated with organ failure and death [47,48]. Bacterial derived lipopolysaccharide (LPS)

drives release of cytokines, such as tumor necrosis-α and interleukins, known as the ‘cytokine storm’.

Additionally, the endothelium becomes activated and increased presence of LPS in the vasculature

is associated with increased expression of endothelial cell adhesion molecules (CAM) P-selectin,

E-selectin, vascular cell adhesion molecule (VCAM-1), intercellular adhesion molecule (ICAM-1)

and platelet and endothelial cell adhesion molecule-1 (PECAM-1) [49]. These adhesion molecules

orchestrate tethering, rolling and firm adhesion of leukocytes on and transmigration across

the endothelium [50]. During sepsis, soluble isoforms of adhesion molecules (sCAMs) accumulate

in the bloodstream due to shedding [51]. Shedding represents removal of CAMs from cell surfaces

GEN

ERA

L INTRO

DU

CTIO

N

15

1by enzymes called sheddases, in particular matrix metalloproteinase-9 (MMP-9) and neutrophil

elastase (NE), released from tertiary granules in neutrophils [51]. Both MMP-9 and NE prepare

the extracellular matrix underlying the endothelium to allow transmigration of leukocytes into

inflammatory sites. The activity of MMP-9 is tightly regulated by sheddase antagonist tissue-

inhibitor of metalloproteinases-1 (TIMP-1) to reduce damage to host-tissues and an increased

TIMP-1/MMP-9 ratio was associated with severity and outcome of sepsis in adults [52,53].

Chapter 5 reviews mechanisms for changes in levels of circulating adhesion molecules and their

sheddases during sepsis and age-dependency of their levels in newborns, children and adults.

For Chapter 6 we applied the concept of simultaneous measurement of circulating adhesion

molecules and their sheddases in a cohort of healthy newborns and newborns with suspected

EOS. We hypothesized that higher circulating levels of adhesion molecules sP-selectin, sE-selectin

sVCAM-1, sICAM-1 and sPECAM-1, coincide with higher levels of sheddases MMP-9 and NE, and

sheddase antagonist TIMP-1 in newborns with culture proven EOS versus healthy controls.

ENDOTHELIAL INTEGRITY DURING EARLY ONSET SEPSIS: THE ANGIOPOIETINSEndothelial integrity is maintained by the Angiopoietin/Tie2 Receptor Tyrosine Kinase - system,

which consists of the endothelial restricted receptor Tie-2 and its ligands Angiopoietin (Ang)-1

and Ang-2 [54]. In health, Ang-1 is present in human serum at higher levels than Ang-2 and

promotes endothelial stability through continuous endothelial Tie-2 receptor phosphorylation

[55]. Inflammation leads to higher circulating levels of Ang-2 that is being release from endothelial

cells. Circulating Ang-2 dose-dependently inhibits Tie-2 signaling and acts as an antagonist of Ang/

Tie-2, driving vascular permeability. Emerging clinical evidence indicates a positive correlation of

high Ang-2 levels, and subsequent high Ang-2/Ang-1 ratio with presence, severity, and outcome

of pediatric and adult sepsis [56,57]. It was recently suggested that the Angiopoietins may be

relevant as biomarkers of EOS [58]. Additionally, investigating the dynamics of Ang-1 and Ang-2

in healthy and infected newborns may unravel changes in their levels during EOS. In Chapter 7,

these changes are explored in a large cohort of healthy newborns and newborns with suspected

and culture proven EOS. For this study, we hypothesized that low Ang-1 and high Ang-2 levels are

associated with presence of bacterial culture positive EOS.

NOVEL ASPECTS OF NEUTROPHILS IN SEPSISManual microscopic analysis of neutrophils and their counts have been part of the clinical

assessment of bacterial infection for over a century [59]. However, manual analysis of counts

and morphology is time consuming, requires experienced laboratory technicians, and lacks

reproducibility. Novel methods allow for measurement of several aspects of neutrophils, in

particular morphology, mechanics and motility. Flow-based automated hematology analysers

(AHAs) are able to determine leukocyte subsets and different granulocyte fractions [42].

Additionally, recent developments in the performance of these AHAs have enabled measurement

of neutrophil size and scatter properties and determination of neutrophil cell surface markers with

GEN

ERA

L INTRO

DU

CTIO

N

16

1immunofluorescence, each with their own sensitivity for presence of sepsis in patients. Chapter 8

and Chapter 9 discuss basic and clinical aspects of neutrophil morphology, mechanics and motility

during sepsis, along with current evidence and future possibilities for the use of these parameters

into the management of sepsis.

GEN

ERA

L INTRO

DU

CTIO

N

17

1REFERENCES1. Simonson KA, Anderson-Berry AL, Delair SF,

Davies HD. Early-onset neonatal sepsis. Clin

Microbiol Rev 2014, 27(1):21-47.

2. Schrag SJ, Farley MM, Petit S, Reingold A, Weston

EJ, Pondo T, Hudson Jain J, Lynfield. Epidemiology

of Invasive Early-Onset Neonatal Sepsis, 2005 to

2014. Pediatrics 2016, 138(6), pii: e20162013.

3. van Herk W, Stocker M, van Rossum AM.

Recognising early onset neonatal sepsis: an

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77-82.

4. Cohen-Wolkowiez M, Moran C, Benjamin DK,

Cotten CM, Clark RH, Benjamin DK, Jr, Smith PB.

2009. Early and late onset sepsis in late preterm

infants. Pediatr. Infect. Dis. J. 2009, 28:1052–1056.

5. Stoll BJ, Hansen NI, Sanchez PJ, Faix RG,

Poindexter BB, Van Meurs, KP, Bizzarro MJ,

Goldberg RN, Frantz ID, III, Hale EC, Shankaran

S, Kennedy K, Carlo WA, Watterberg KL, Bell

EF, Walsh MC, Schibler K, Laptook AR, Shane

AL, Schrag SJ, Das A, Higgins RD, Eunice

Kennedy Shriver National Institute of Child

Health and Human Development Neonatal

Research Network. Early onset neonatal sepsis:

the burden of group B streptococcal and E. coli

disease continues. Pediatrics 2011, 127:817–826.

6. Weston EJ, Pondo T, Lewis MM, Martell-Cleary

P, Morin C, Jewell B, Daily P, Apostol M, Petit

S, Farley M, Lynfield R, Reingold A, Hansen NI,

Stoll BJ, Shane AJ, Zell E, Schrag SJ.The burden

of invasive early-onset neonatal sepsis in

the United States, 2005-2008. Pediatr Infect

Dis J 2011, 30(11):937-41.

7. Schrag SJ, Verani JR. Intrapartum antibiotic

prophylaxis for the prevention of perinatal

group B streptococcal disease: experience

in the United States and implications for

a potential group B streptococcal vaccine.

Vaccine 2013, 31(4):D20-6.

8. Bauserman MS, Laughon MM, Hornik CP, Smith

PB, Benjamin DK Jr, Clark RH, Engmann C,

Cohen-Wolkowiez M. Group B Streptococcus

and Escherichia coli infections in the intensive

care nursery in the era of intrapartum antibiotic

prophylaxis. Pediatr Infect Dis J 2013, 32(3):208-12.

9. Stoll BJ. Early-onset neonatal sepsis: a continuing

problem in need of novel prevention strategies.

Pediatrics 2016, 138(6): e20163038.

10. Ohlsson A, Shah VS. Intrapartum antibiotics for

known maternal Group B streptococcal colonization.

Cochrane Database Syst Rev 2014, (6):CD007467.

11. Li S, Huang J, Chen Z, Guo D, Yao Z, Ye X.

Antibiotic Prevention for Maternal Group B

Streptococcal Colonization on Neonatal GBS-

Related Adverse Outcomes: A Meta-Analysis.

Front Microbiol 2017, 8:374.

12. Dangor Z, Cutland CL, Izu A, Kwatra G, Trenor

S, Lala SG, Madhi SA. Temporal Changes in

Invasive Group B Streptococcus Serotypes:

Implications for Vaccine Development.

PLoS One 2016, 11(12):e0169101.

13. Chan GJ, Lee AC, Baqui AH, Tan J, Black RE.

Prevalence of early-onset neonatal infection

among newborns of mothers with bacterial

infection or colonization: a systematic review

and meta-analysis. BMC Infect Dis 2015, 15:118.

14. Le Doare K, Heath PT. An overview of global

GBS epidemiology. Vaccine 2013, 28:31(4):7-12.

15. Edmond KM, Kortsalioudaki C, Scott S, Schrag

SJ, Zaidi AK, Cousens S, Heath PT. Group B

streptococcal disease in infants aged younger

than 3 months: systematic review and meta-

analysis. Lancet 2012, 379(9815):547-56.

16. Sinha A, Russell LB, Tomczyk S, Verani JR, Schrag

SJ, Berkley JA, Mohammed M, Sigauque B, Kim

SY; GBS Vaccine Cost-Effectiveness Analysis in

Sub-Saharan Africa Working Group. Disease

Burden of Group B Streptococcus Among

Infants in Sub-Saharan Africa: A Systematic

Literature Review and Meta-analysis. Pediatr

Infect Dis J 2016, 35(9):933-42.

17. Lawn JE, Cousens S, Zupan J; Lancet

Neonatal Survival Steering Team. 4 million

neonatal deaths: when? Where? Why?

Lancet 2005, 365(9462):891-900.

18. Chow S, Chow R, Popovic M, et al. A Selected

Review of the Mortality Rates of Neonatal Intensive

Care Units. Front Public Health 2015, 7;3:225

19. Quan V, Verani JR, Cohen C, von Gottberg

A, Meiring S, Cutland CL, Schrag SJ, Madhi

GEN

ERA

L INTRO

DU

CTIO

N

18

1SA. Invasive Group B Streptococcal Disease

in South Africa: Importance of Surveillance

Methodology. PLoS One 2016, 11(4):e0152524.

20. Dagnew AF, Cunnington MC, Dube Q, Edwards

MS, French N, Heyderman RS, Madhi SA,

Slobod K, Clemens SA. Variation in reported

neonatal group B streptococcal disease

incidence in developing countries. Clin Infect

Dis 2012, 55(1):91-102.

21. Census Statistics Suriname, 2012. http://www.

statistics-suriname.org

22. Mazoembaks N. Verzwegen werk van P.C.

Flu: kindersterfte in Suriname, een erfenis uit

de slavernij. Uitgeverij De Woordenwinkel,

Zierikzee 2014, pp 48 and 80.

23. Zijlmans W, Hindori-Mohangoo A.

Determinants of neonatal mortality in

Suriname: preliminary findings from a perinatal

and infant mortality survey. Ann of Glob

Health 2015, 81(1): 121-122.

24. WHO data, 2015. http://www.who.int

25. Bekker V, Bijlsma MW, van de Beek D, Kuijpers

TW, van der Ende A. Incidence of invasive group

B streptococcal disease and pathogen genotype

distribution in newborn babies in the Netherlands

over 25 years: a nationwide surveillance study.

Lancet Infect Dis 2014, 14(11):1083-9.

26. van den Anker JN. How to optimize

the evaluation and use of antibiotics in

neonates. Early Hum Dev 2014, 90(1):S10-2.

27. Canty JB, Wozniak PS, Sanches PJ. Prospective

surveillance of antibiotic use in the neonatal

intensive care unit: results from the SCOUT

study. Pediatr Infect Dis J 2015;34:267-72.

28. Cantey JB, Wozniak PS, Pruszynski JE, Sánchez

PJ. Reducing unnecessary antibiotic use in

the neonatal intensive care unit (SCOUT):

a prospective interrupted time-series study.

Lancet Infect Dis 2016, 16(10):1178-84.

29. Cotten CM. Adverse consequences of

neonatal antibiotic exposure. Curr Opin

Pediatr 2016, 28(2):141-9.

30. Langdon A, Crook N, Dantas G. The effects

of antibiotics on the microbiome throughout

development and alternative approaches for

therapeutic modulation. Genome Med, 2016, 8(1):39.

31. Cotten CM, Taylor S, Stoll B, Goldberg RN,

Hansen NI, Sánchez PJ, Ambalavanan N,

Benjamin DK; NICHD Neonatal Research

Network. Prolonged duration of initial

empirical antibiotic treatment is associated

with increased rates of necrotizing

enterocolitis and death for extremely low birth

weight infants. Pediatrics 2009, 123(1):58–66.

32. Kuppala VS, Meinzen-Derr J, Morrow AL,

Schibler KR. Prolonged initial empirical

antibiotic treatment is associated with

adverse outcomes in premature infants. J

Pediatr 2011, 159(5):720–725.

33. Escobar GJ, Puopolo KM, Wi S, et al. Stratification

of risk of early-onset sepsis in newborns ≥ 34

weeks’ gestation. Pediatrics. 2014;133:30–36.

34. Shakib J, Buchi K, Smith E, et al. Management

of newborns born to mothers with

chorioamnionitis: Is it time for a kinder, gentler

approach? Acad. Pediatr, 2015;15:340–344.

35. Kerste M, Corver J, Sonnevelt MC, van Brakel

M, van der Linden PD, M Braams-Lisman BA,

Plötz FB. Application of sepsis calculator in

newborns with suspected infection. J Matern

Neonatal Med 2016;7058:1–6.

36. Pepys MB, Hirschfield GM. C-reactive protein:

a critical update. J Clin Invest 200, ;111:1805–1812.

37. Hofer N, Zacharias E, Müller W, Resch B. An

update on the use of C-reactive protein in

early-Onset neonatal sepsis: Current insights

and new tasks. Neonatology 2012, 102:25–36.

38. Mukherjee A, Davidson L, Anguvaa L, Duffy

DA, Kennea N. NICE neonatal early onset

sepsis guidance: greater consistency, but more

investigations, and greater length of stay. Arch

Dis Child Fetal Neonatal Ed 2015, 100(3):F248-9.

39. Newman TB, Puopolo KM, Wi S, Draper D,

Escobar GJ. Interpreting complete blood

counts soon after birth in newborns at risk for

sepsis. Pediatrics 2010, 126:903–90.

40. MacQueen BC, Christensen RD, Yoder BA, Henry

E, Baer VL, Bennett ST, Yaish HM. Comparing

automated vs manual leukocyte differential

counts for quantifying the ‘left shift’ in the blood

of neonates. J Perinatol 2016, 36(10):843-8.

41. Wiland EL, Sandhaus LM, Georgievskaya Z, Hoyen

CM, O’Riordan MA, Nock ML. Adult and child

GEN

ERA

L INTRO

DU

CTIO

N

19

1automated immature granulocyte norms are

inappropriate for evaluating early-onset sepsis in

newborns. Acta Paediatr 2014;103(5):494-7.

42. Mikhael M, Brown LS, Rosenfeld CR. Serial

neutrophil values facilitate predicting

the absence of neonatal early-onset sepsis. J

Pediatr 2014;164(3):522-8.

43. van Herk W, el Helou S, Janota J, Hagmann C,

Klingenberg C, Staub E, Giannoni E, Tissieres P,

Schlapbach LJ, van Rossum AM, Pilgrim SB, Stocker

M. Variation in Current Management of Term and

Late-preterm Neonates at Risk for Early-onset

Sepsis: An International Survey and Review of

Guidelines. Pediatr Infect Dis J 2016, 35(5):494-500.

44. Chauhan N, Tiwari S, Jain U. Potential

biomarkers for effective screening of neonatal

sepsis infections: An overview. Microb

Pathog 2017, 107:234-242.

45. Stocker M, van Herk W, El Helou S, Dutta

S, Fontana MS, Schuerman FABA, van den

Tooren-de Groot RK, Wieringa JW, Janota J, van

der Meer-Kappelle LH, Moonen R, Sie SD, de

Vries E, Donker AE, Zimmerman U, Schlapbach

LJ, de Mol AC, Hoffman-Haringsma A, Roy M,

Tomaske M, Kornelisse RF, van Gijsel J, Visser

EG, Willemsen SP, van Rossum AMC; NeoPInS

Study Group. Procalcitonin-guided decision

making for duration of antibiotic therapy in

neonates with suspected early-onset sepsis:

a multicentre, randomised controlled trial

(NeoPIns). Lancet 2017, pii. 6736(17)31444-7.

46. Aird WC. The role of the endothelium in

severe sepsis and multiple organ dysfunction

syndrome. Blood 2003, 101(10):3765-77.

47. Brown KA, Brain SD, Pearson JD, Edgeworth

JD, Lewis SM, Treacher DF. Neutrophils in

development of multiple organ failure in

sepsis. Lancet 2006, 368:157-69.

48. Shapiro NI, Schuetz P, Yano K, Sorasaki M,

Parikh SM, Jones AE, Trzeciak S, Ngo L, Aird WC.

The association of endothelial cell signalling,

severity of illness, and organ dysfunction in

sepsis. Crit Care 2010, 14(5):R182.

49. Kümpers P, van Meurs M, David S, Molema G,

Bijzet J, Lukasz A, Biertz F, Haller H, Zijlstra JG.

Time course of angiopoietin-2 release during

experimental human endotoxemia and sepsis.

Crit Care 2009, 13(3):R64.

50. Ley K, Laudanna C, Cybulsky MI, Nourshargh

S. Getting to the site of inflammation:

the leukocyte adhesion cascade updated. Nat

Rev Immunol 2007, 7(9):678-89.

51. Garton KJ, Gough PJ, Raines EW. Emerging roles

for ectodomain shedding in the regulation

of inflammatory responses. J Leukoc

Biol 2006, 79(6):1105-16.

52. Lorente L, Martín MM, Solé-Violán J, Blanquer

J, Labarta L, Díaz C, Borreguero-León JM,

Orbe J, Rodríguez JA, Jiménez A, Páramo JA.

Association of sepsis-related mortality with

early increase of TIMP-1/MMP-9 ratio. PLoS

One 2014, 9(4):e94318.

53. Wang M, Zhang Q, Zhao X, Dong G, Li C.

Diagnostic and prognostic value of neutrophil

gelatinase-associated lipocalin, matrix

metalloproteinase-9, and tissue inhibitor

of matrix metalloproteinases-1 for sepsis in

the Emergency Department: an observational

study. Crit Care 2014, 18(6):634.

54. van Meurs M, Kümpers P, Ligtenberg JJ,

Meertens JH, Molema G, Zijlstra JG. Bench-to-

bedside review: Angiopoietin signalling in critical

illness - a future target? Crit Care 2009, 13(2):207.

55. Parikh SM. Dysregulation of

the angiopoietin-Tie-2 axis in sepsis and ARDS.

Virulence 2013, 4(6):517-24.

56. Fang Y, Li C, Shao R, Yu H, Zhang Q, Zhao L.

Prognostic significance of the angiopoietin-2/

angiopoietin-1 and angiopoietin-1/Tie-2 ratios

for early sepsis in an emergency department.

Crit Care 2015, 14(19):367.

57. Mussap M, Cibecchini F, Noto A, Fanos V. In search

of biomarkers for diagnosing and managing

neonatal sepsis: the role of angiopoietins. J

Matern Fetal Neonatal Med 2013, 26(2):24-6.

58. Giuliano JS Jr, Tran K, Li FY, Shabanova V,

Tala JA, Bhandari V. The temporal kinetics of

circulating angiopoietin levels in children with

sepsis. Pediatr Crit Care Med 2014, 15(1):e1-8.

59. Cornbleet PJ. Clinical utility of the band count.

Clin Lab Med 2002, 22(1):101-36.

I Epidemiology of Early Onset Sepsis in Suriname

2 Improved Referral and Survival of Newborns after Scaling Up of

Intensive Care in Suriname

Rens Zonneveld, Natanael Holband, Anna Bertolini, Francesca Bardi, Neirude Lissone, Peter Dijk,

Frans B. Plötz, Amadu Juliana

BMC Pediatrics, Accepted for Publication

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

28

2

ABSTRACTBackground

Scaling up neonatal care facilities in developing countries can improve survival of newborns.

Recently, the only tertiary neonatal care facility in Suriname transitioned to a modern environment

in which interventions to improve intensive care were performed. This study evaluates impact of

this transition on referral pattern and outcomes of newborns.

Methods

A retrospective chart study amongst newborns admitted to the facility was performed and

outcomes of newborns between two 9-month periods before and after the transition in March

2015 were compared.

Results

After the transition more intensive care was delivered (RR 1.23; 95% CI 1.07-1.42) and more outborn

newborns were treated (RR 2.02; 95% CI 1.39-2.95) with similar birth weight in both periods (P=0.16).

Mortality of inborn and outborn newborns was reduced (RR 0.62; 95% CI 0.41-0.94), along with

mortality of sepsis (RR 0.37; 95% CI 0.17-0.81) and asphyxia (RR 0.21; 95% CI 0.51-0.87). Mortality

of newborns with a birth weight <1000 grams (34.8%; RR 0.90; 95% CI 0.43-1.90) and incidence

of sepsis (38.8%, 95% CI 33.3-44.6) and necrotizing enterocolitis (NEC) (12.5%, 95% CI 6.2-23.6)

remained high after the transition.

Conclusions

After scaling up intensive care at our neonatal care facility more outborn newborns were admitted

and survival improved for both in- and outborn newborns. Challenges ahead are sustainability,

further improvement of tertiary function, and prevention of NEC and sepsis.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

29

2

BACKGROUNDNeonatal mortality in developing countries continues to be a chief global health challenge [1,2].

A recent global report indicates that over 40% reduction of neonatal mortality can be achieved

by implementation of institutional care in lower resource countries [3]. In particular, local or

regional neonatal care facilities with integrated availability of perinatal and neonatal intensive

care can reduce mortality [4]. For example, newborns born in a rural hospital featuring a neonatal

intensive care unit (NICU) in Uganda were almost twice as likely to survive than those born outside

[5]. Moreover, introduction of a neonatal care facility in a low-income district in India reduced

neonatal mortality rate (NMR) by 21% after the first two years [6]. Improving interventions within

existing neonatal care facilities (e.g., training of personnel, refurbishment, infection prevention)

can improve mortality and enhance tertiary function for newborns in need of intensive care [6-9].

In Suriname NMR in 2009 was 16.0 per 1000 live births. However, detailed data on demographics

and outcomes of newborns are lacking. In 2008 the neonatal care facility at the Academic Hospital

Paramaribo (AHP), which also incorporated the first and only NICU in Suriname, opened its doors.

The ability to treat premature and critically ill newborns was an important step towards reducing

mortality. At the end of March 2015 the facility moved to a new and modern environment. This

transition solidified availability of neonatal intensive care in Suriname with reinforcement and

training of personnel, new equipment, continuous availability of supplies, and protocol-based

care. Since this facility is the only referral center for newborns requiring intensive care in Suriname,

morbidity and mortality of newborns treated here reflect their outcomes at the national level.

Therefore, as a benchmark for future investigations, we developed a registry to describe

demographics and outcomes of newborns admitted to the neonatal care facility. Additionally, to

evaluate the impact of improvements we compare referral pattern, mortality and morbidity of

newborns treated in periods before and after the transition. Ultimately, this could lead to better

prospective registry and care for critically ill newborns in Suriname.

METHODSStudy Design

We performed a retrospective (pre-and post transition) study in the neonatal care facility of

the AHP during the periods July 1st 2014 to March 29th 2015 (Period 1) and March 31st to December

31st 2015 (Period 2). The impact of the transition was described by analyzing demographics and

outcomes of all inborn and outborn newborns admitted within these two periods. Excluded were

newborns whom were treated in both periods and of whom insufficient information (i.e., no or

incomplete paper charts) was available to confirm outcomes. We received a waiver from our

institutional ethical board.

Setting and Interventions

Suriname is a small middle-income country with a multiethnic society and has an annual birth rate

of about 10,000 births. Over 90% of births take place at delivery rooms of one of four hospitals

situated in Suriname’s capital Paramaribo (inhabited by more than half of Suriname’s population).

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

30

2

About 30% of births take place at the delivery room of the AHP. The neonatal care facility at

the AHP serves as the only referral hospital for critically ill newborns. Since the opening in 2008,

between 350-400 newborns are treated each year in one room with 12 beds, with NICU capacity

operating at Level III [9]. Newborns are generally only actively treated with a birth weight (BW) ≥

750 grams and/or gestational age (GA) ≥ 27 weeks.

On March 30th 2015 the facility moved to a completely new, modern and spacious environment

with central climate control and new equipment (i.e., ventilators, incubators, air-humidifiers,

ultrasound machines and multi-parameter monitors). Capacity for mechanical ventilation and

continuous positive airway pressure (CPAP) was doubled. The NICU (6 beds), high care (HC)

(6 beds), and medium care (MC) (4 beds) capacity in the new facility remained the same until

February 2016 (when a separate space for the MC was opened and the NICU capacity increased to

10 beds).

Total expense for the new building and equipment was 2.6 million US dollars. Funds were

collected from kind donations from governmental and private organizations and from Surinamese

companies. Since there were no architects or contractors available within Suriname with experience

in designing a NICU level neonatal care facility, we relied on guidelines from developed countries

and local creativity and practical experience to realize the project within budget, without the need

for expensive consultants. For example, one of the savings came from using venturi mechanism

based suction devices powered by compressed air, avoiding the need for a separate central

vacuum system.

Admission criteria remained the same. Obstetric nurses were trained in neonatal life support

and the number of residents in the obstetric and pediatric department was increased. For both

day and evening shifts a separate resident was assigned to the NICU exclusively. Shortly before

the transition, nurses were trained in intensive neonatal care and their number was expanded

to 1 per 3 or 4 beds. New charts for vital signs, ventilation settings, and fluid management were

implemented. A breast-feeding and nutrition program was started to help reduce cases of

necrotizing enterocolitis (NEC) and mothers were allowed at the bedside twice as long as before.

Systematic infection prevention (i.e., stringent guidelines and more facilities for hand washing,

providing of patient specific (disposable) materials, Extended Spectrum Beta-Lactamase (ESBL)

outbreak control) was enforced.

Data Collection and Analysis

Data were collected from paper medical records on maternal, obstetric and perinatal history,

birth location, reason for admission, hospital course, and outcomes. A single major cause of death

was determined. For each included newborn we determined the highest level of care during

their stay by assigning criteria for NICU, HC or MC retrospectively according to local protocol

(Supplemental Table 1). Primary outcome was mortality: NMR at the AHP and at the neonatal care

facility divided in early (i.e., in-hospital death before 7 days of life) and late (i.e., in-hospital death

of at term newborns after 7 days of life), GA-specific mortality, BW-specific mortality, and cause-

specific mortality. Secondary outcomes were highest level of care, respiratory treatments (CPAP,

mechanical ventilation, surfactant), use of antibiotics, development of respiratory complications,

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

31

2

i.e., pneumothorax, bronchopulmonary dysplasia (BPD; i.e., oxygen dependence > 28 days of age),

ventilator-associated pneumonia (VAP; i.e., positive tracheal aspirate culture after ventilation),

development of NEC and sepsis (i.e., early (<72 hours after birth) and late (>72 hours after birth)

onset clinical (i.e., clinical suspicion, treated with antibiotics for 7 days, raised c-reactive protein

levels)) and blood culture positive sepsis, blood and ESBL culture results, and duration of stay.

Statistical Analysis

Incidence rates and epidemiological determinants were calculated for the inclusion period.

Categorical variables are presented as numbers and percentages with 95% confidence intervals

(CI) and continuous variables as means with standard deviations (SD) or, if not normally distributed,

as medians with ranges. Continuous variables were compared with a student t-test and categorical

variables were compared with Chi-Square. Relative risk (RR) and 95% CI were calculated.

P-values < 0.05 were considered statistically significant.

RESULTSDemographics and Referral

A total of 626 newborns were treated at the neonatal care facility of whom 601 (320 before and

281 after the transition) were included (Table 1). Overall demographics were comparable between

both periods, with similar percentages of missing data, showing high prevalence of (antenatal) risk

factors for mortality and morbidity (Table 1). In period 2 significantly more outborn newborns (RR

2.02; 95% CI 1.39-2.95; P<0.001) were treated with similar mean birthweight (2183 ± 845 grams vs.

1915 ± 990 grams; P=0.16). Prematurity was the main reason for admission for all inborn (48.3%; 95%

CI 44.0-52.7) and outborn (66.0%; 95% CI 56.3-74.5) newborns, followed by respiratory distress and

suspected infection (Table 1).

Mortality

NMR of inborn newborns born at the AHP was lower in period 2 (P=0.02) (Table 2). After

the transition, reduction in mortality was greatest in newborns treated at NICU level care (P<0.01),

with a GA above 28 weeks (RR 0.42; 95% CI 0.25-0.72; P=0.002), and outborn newborns (P=0.02).

A trend in decrease in mortality was observed in late mortality (P=0.06), inborn newborns (P=0.07),

and in newborns with a birth weight (BW) above 1500 grams (P=0.07). A significant reduction in

mortality was observed in cases of sepsis (P=0.01) and perinatal asphyxia (P=0.03). Sepsis was

the main cause of death in period 1 (34.5%; 95% CI 23.4-47.7), and second in period 2 (26.7%; 95%

CI 14.2-44.4). For newborns with a BW<1000 grams late-onset sepsis was the main cause of death

in both periods (44.8%; 95% CI 28.4-62.5).

Treatments and Morbidity

Based on our criteria (Supplemental Table 1) significantly more NICU level care was given in

period 2 (P<0.01) (Table 3a). More mechanical ventilation and surfactant were applied after

the transition. No difference in prevalence of VAP or pneumothorax was observed and there was

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

32

2

Table 1. Demographics of newborns admitted to the neonatal care facility before and after the transition

Period 1

(July 2014-March 2015)

Period 2

(April 2015-December 2015)

N % (95% CI) N % (95% CI)

Live births Total at AHP 2353 1972

Admissions to

facility

Total

Included

Inborn

Outborn2

331

320

284

36

96.7

88.7 (84.8-91.8)

11.3 (8.2-15.2)

295

281

217

64

95.3

77.2 (72.0-81.7)

22.8 (18.3-28.0)

Maternal age

(Years)

<20

20-34

≥35

Missing

54

168

46

52

16.9 (13.2-21.4)

52.5 (47.0-57.9)

14.4 (11.0-18.6)

16.3

36

140

24

81

12.8 (9.4-17.2)

49.8 (44.0-55.6)

8.5 (5.8-12.4)

28.8

Pregnancy HIV

Diabetes

PIH / Preeclampsia

Antenatal steroids3

Infection risk4

6

18

60

47

47

1.9 (0.9-4.0)

5.6 (3.6-8.7)

18.8 (14.9-23.4)

14.7 (11.2-19.0)

14.7 (11.2-19.0)

2

20

62

55

38

0.7 (0.2-2.6)

7.1 (4.7-10.7)

22.1 (17.6-27.3)

19.6 (15.4-24.6)

13.5 (10.0-18.0)

Mode of delivery Vaginal

Caesarean section

Missing

187

105

28

58.4 (53.0-63.7)

32.8 (27.9-38.1)

8.8

167

94

20

59.4 (53.6-65.0)

33.5 (28.2-39.2)

7.1

Sex Male

Female

162

158

50.6 (45.2-56.1)

49.4 (43.9-54.8)

155

126

55.2 (49.3-60.9)

44.8 (39.1-50.7)

Gestational age

(Weeks)

<28

28-32

33-36

≥37

Missing

16

48

114

132

10

5.0 (3.1-8.0)

15.0 (11.5-19.3)

35.6 (30.6-41.0)

41.3 (36.0-46.7)

3.1

13

47

100

110

11

4.6 (2.7-7.8)

16.7 (12.8-21.5)

35.6 (30.2-41.3)

39.1 (33.6-45.0)

3.9

Birth weight

(Grams)

<1000

≥1000-1499

≥1500

Missing

26

48

242

4

8.1 (5.6-11.6)

15.0 (11.5-19.3)

75.6 (70.6-80.0)

1.3

23

33

221

4

8.2 (5.5-12.0)

11.7 (8.5-16.0)

78.6 (73.5-83.0)

1.4

Apgar Score at 5’ <5

Missing

24

45

7.5 (5.1-10.9)

14.1

7

47

2.5 (1.2-5.1)

16.7 (12.8-21.5)

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

33

2

a trend in increases incidence of BPD (P=0.07) (Table 3b). Grade 2 or higher NEC was present at

high incidence in newborns with a BW<1500 grams in both periods (5.4% and 12.5%, respectively).

Sepsis (either early or late-onset) was prevalent in over 30% of patients in both periods, of which

half was LOS. During both periods, outbreaks with ESBL bacteria led to a significant prevalence of

ESBL positive cultures.

Table 1. (continued)

Period 1

(July 2014-March 2015)

Period 2

(April 2015-December 2015)

N % (95% CI) N % (95% CI)

Ethnicity Maroon

Creole

Hindo-Surinamese

Javanese

Amerindian

Chinese

Other5

Missing

87

85

59

15

10

2

31

31

27.2 (22.6-32.3)

26.2 (22.0-31.7)

18.4 (14.6-23.1)

4.7 (2.9-7.6)

3.1 (1.7-5.7)

0.6 (0.2-2.2)

9.7 (6.9-13.4)

9.7

72

72

55

21

7

2

32

20

25.6 (20.9-31.0)

25.6 (20.9-31.0)

19.6 (15.4-24.6)

7.5 (4.9-11.2)

2.5 (1.2-5.1)

0.7 (0.2-2.6)

11.4 (8.2-15.6)

7.1

Initial reason for

admission1

Prematurity

Respiratory distress6

Suspected infection7

Perinatal asphyxia8

Congenital malformations9

Other10

152

119

91

39

42

71

47.5 (42.1-53.0)

37.2 (32.1-42.6)

28.4 (23.8-33.6)

12.2 (9.0-16.2)

13.1 (9.9-17.3)

22.2 (18.0-27.1)

148

122

97

30

35

49

52.7 (46.8-58.4)

43.4 (37.7-49.3)

34.5 (29.2-40.3)

10.7 (7.6-14.8)

12.5 (9.1-16.8)

17.4 (13.4-22.3)

AHP = Academic Hospital Paramaribo; NICU = neonatal intensive care unit; HC = high care; MC = medium care; PIH =

pregnancy-induced hypertension; RDS = respiratory distress syndrome. 1 Newborns could have more than one reason for admission.2 Includes: delivery rooms of four other hospitals in Paramaribo and one other hospital in Nickerie, birth clinics in rural and

interior parts of Suriname, and home births.3Administered in two doses of dexamethasone in the case of suspected premature birth before GA of 34 weeks.4 Includes: premature rupture of membranes (PROM), intrapartum fever and/or antibiotics, positive maternal Group-B

streptococcus culture.

5 Includes: Caucasian, Brazilian, or mixed,6 Includes: neonatal respiratory distress syndrome, congenital pneumonia, pulmonary hemorrhage, pneumothorax,

meconium aspiration syndrome, and transient neonatal tachypnea.7 Includes: newborns defined with clinical symptoms of infection by admitting physician.8 Includes: asphyxia defined by admitting physician (e.g., in the case of either need for resuscitation or Apgar <5 beyond

5 minutes; lactate acidosis with base excess <16; coma or seizures after birth; findings with cerebral ultrasound such

as edema).9 Includes: diaphragmatic hernia, congenital heart defects, gastro-intestinal anomalies and neurological malformations.10 Includes: hypoglycemia, dysmaturity, jaundice, and social indications.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

34

2

Tab

le 2

. Mo

rtal

ity

of n

ewb

orn

s tr

eate

d at

the

faci

lity

bef

ore

and

aft

er th

e tr

ansi

tio

n Peri

od

1 (

N=3

20)

(Jul

y 20

14-M

arch

20

15)

Peri

od

2 (

N=2

81)

(Ap

ril 2

015

-Dec

emb

er 2

015

)

Rel

ativ

e R

isk

(95%

CI)

P-va

lue

N%

N%

Ove

rall

mo

rtal

ity

Tota

l at

AH

P (p

er 10

00

live

bir

ths)

1

Tota

l at

faci

lity

To

tal e

arly

neo

nata

l mo

rtal

ity

To

tal l

ate

neo

nata

l mo

rtal

ity

In

bo

rn

O

utb

orn

N

ewb

orn

s w

ith

NIC

U le

vel c

are

23.4

55/3

20

29/3

20

26/3

20

42/2

84

13/3

6

52/1

59

17.2

9.1

8.1

14.8

36.1

32.7

13.2

30/2

81

18/2

81

12/2

81

20/2

17

10/6

4

29/1

72

10.7

6.4

4.3

9.2

15.6

16.9

0.5

6 (0

.36-

0.9

0)

0.6

2 (0

.41-

0.9

4)

0.7

0 (

0.4

0-1

.24)

0.5

3 (0

.27-

1.0

2)

0.6

2 (0

.38-

1.0

3)

0.4

3 (0

.21-

0.8

9)

0.5

2 (0

.35-

0.7

7)

0.0

2

0.0

2

0.2

3

0.0

6

0.0

7

0.0

2

<0.0

1

Ges

tati

ona

l age

-sp

ecifi

c

mo

rtal

ity

<28

wee

ks

28-3

2 w

eeks

33-3

6 w

eeks

≥37

wee

ks

Mis

sing

6/16

12/4

8

14/1

14

20/1

32

3

37.5

25.0

12.3

15.2

8/13

5/47

4/10

0

8/11

0

5

61.5

10.6

4.0

7.3

1.64

(0

.76-

3.53

)

0.4

3 (0

.16-1

.11)

0.3

3 (0

.11-0

.96)

0.4

8 (0

.22-

1.0

5)

0.2

0

0.0

8

0.0

4

0.0

7

Birt

h w

eigh

t-sp

ecifi

c m

ort

alit

y<1

00

0 g

≥10

00

-14

99 g

≥150

0 g

M

issi

ng

10/2

6

13/4

8

30/2

42

2

38.5

27.1

12.4

8/23

6/33

16/2

21

0

34.8

18.2

7.2

0.9

0 (

0.4

3-1.9

0)

0.6

7 (0

.28-

1.59

)

0.5

8 (0

.33-

1.0

4)

0.7

9

0.3

6

0.0

7

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

35

2

Tab

le 2

. (co

ntiu

ed)

Peri

od

1 (

N=3

20)

(Jul

y 20

14-M

arch

20

15)

Peri

od

2 (

N=2

81)

(Ap

ril 2

015

-Dec

emb

er 2

015

)

Rel

ativ

e R

isk

(95%

CI)

P-va

lue

Cau

se-s

pec

ific

mo

rtal

ity

Seps

is2

Ea

rly-

ons

et s

epsi

s

La

te-o

nset

sep

sis

Peri

nata

l asp

hyxi

a

Prem

atur

ity

com

plic

atio

n3

Co

ngen

ital

mal

form

atio

ns4

Oth

er5

19/9

6

10/4

4

9/52

12/3

8

7/15

7

12/4

2

5

19.8

22.7

17.3

31.6

4.5

28.6

8/10

9

3/59

5/50

2/30

5/14

8

9/35

6

7.3

5.1

10.0

6.7

3.4

25.7

0.3

7 (0

.17-0

.81)

0.2

2 (0

.07-

0.7

7)

0.5

8 (0

.21-

1.61

)

0.2

1 (0

.51-

0.8

7)

0.7

6 (0

.25-

2.34

)

0.9

0 (

0.4

3-1.

88)

0.0

1

0.0

2

0.2

9

0.0

3

0.6

3

0.7

8

AH

P =

Aca

dem

ic H

osp

ital

Par

amar

ibo

; NIC

U =

neo

nata

l int

ensi

ve c

are

unit

;1 In

clud

ing

dea

ths

at t

he d

eliv

ery

roo

m (

13 b

efo

re a

nd 6

aft

er t

he t

rans

itio

n).

2 In

clud

es: n

ewb

orn

s w

ith

clin

ical

sus

pici

on,

tre

ated

wit

h an

tibi

oti

cs fo

r 7

day

s, r

aise

d c

-rea

ctiv

e pr

ote

in le

vels

, and

po

siti

ve b

loo

d c

ultu

re.

3 In

clud

es: r

espi

rato

ry in

suffi

cien

cy o

r pn

eum

oth

ora

x w

ith

RDS

and

ext

rem

e pr

emat

urit

y, n

ecro

tizi

ng e

nter

oco

litis

; int

rave

ntri

cula

r he

mo

rrha

ge.

4 In

clud

es: d

iaph

ragm

atic

her

nia,

co

ngen

ital

hea

rt d

efec

ts, g

astr

o-i

ntes

tina

l ano

mal

ies

and

neu

rolo

gica

l mal

form

atio

n.5 In

clud

es: p

ersi

sten

t pu

lmo

nary

hyp

erte

nsio

n o

f the

neo

nate

(PP

HN

), p

neum

oth

ora

x, c

ard

iac

tam

po

nad

e, a

nd k

erni

cter

us.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

36

2

Table 3a. Trends in treatments at the facility in two time periods

Period 1 (N=320)

(July 2014-

March 2015)

Period 2 (N=281)

(April 2015-

December 2015)Relative Risk

(95% CI) P-valueN % N %

Highest level

of care1

NICU

HC

MC

159

75

86

49.7

23.4

26.9

172

60

49

61.2

21.4

17.4

1.23 (1.07-1.42)

0.91 (0.68-1.23)

0.65 (0.47-0.87)

<0.01

0.54

<0.01

Respiratory

treatment

CPAP

Mechanical ventilation

Surfactant

100

38

15

31.3

11.9

4.7

106

55

21

37.7

19.6

7.5

1.21 (0.97-1.51)

1.65 (1.13-2.41)

1.59 (0.84-3.03)

0.10

0.01

0.16

Antibiotics

received

Total 173 54.1 170 60.5 1.12 (0.97-1.29) 0.11

NICU = neonatal intensive care unit; HC = high Care; MC = medium Care; CPAP = continuous positive airway pressure.1 Determined with local criteria given in Supplemental Table 1.

Table 3b. Morbidity of newborns treated at the facility in two time periods

Period 1 (N=320)

(July 2014-

March 2015)

Period 2 (N=281)

(April 2015-

December 2015)Relative Risk

(95% CI) P-valueN % N %

Respiratory

morbidity

BPD

VAP

Pneumothorax

4

9

4

1.3

2.8

1.3

10

5

7

3.6

1.8

2.5

2.85 (0.90-8.98)

0.63 (0.21-1.87)

1.99 (0.59-6.74)

0.07

0.41

0.27

NEC1 Total

≥Stage 2

10

4

13.5

5.4

12

7

21.4

12.5

1.59 (0.74-3.40)

2.31 (0.71-7.51)

0.24

0.16

Sepsis2 Total

Positive blood

culture

96

38

30.0

11.9

109

25

38.8

8.9

1.29 (1.03-1.62)

0.75 (0.46-1.20)

0.02

0.24

Positive ESBL

culture3

Total 34 10.6 39 13.9 1.31 (0.85-2.01) 0.22

Duration of

stay (days)

Mean

13

SD

16

Mean

14

SD

18 0.44

BPD = bronchopulmonary dysplasia; VAP = ventilator-associated pneumonia; NEC = necrotizing enterocolitis; ESBL =

extended spectrum beta-lactamase. 1 Calculated for newborns with a birthweight below 1500 grams (N=74 and N=56 in period 1 and period 2, respectively).2 Includes: early and late-onset clinical (i.e., high clinical suspicion, treated with antibiotics for 7 days; raised C-reactive

protein levels) and blood culture positive sepsis.3 Includes: blood and urine cultures and cultures on (tracheal aspirate, skin and anal) swabs, central lines or

ventilation tubes.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

37

2

DISCUSSIONImprovements at the neonatal care facility led to an increase of newborns that received intensive

care with a significant reduction in their mortality. Furthermore, newborns with a GA above 28

weeks and/or BW≥1500 grams showed a significantly reduced mortality rate. A striking reduction

in mortality was seen in cases of perinatal asphyxia and sepsis. In addition, after the transition

a two-fold increase in admission of outborn newborns, with similar demographics and increased

survival rates, was observed. These findings indicate enhanced tertiary function and centralization

of neonatal intensive care in Suriname, which may play a significant role in reducing neonatal

mortality in Suriname.

Other studies performed in developing countries have shown similar patterns in improvement

of mortality after scaling up of neonatal care facilities. Creation of a level II sick newborn care unit

(SNCU) (i.e., with introduction of bed warmers and central oxygen) in a district hospital in India led

to a significant reduction of regional NMR of mostly newborns with a BW<1500 grams [6]. Another

pre-and-post intervention study in India showed that basic interventions (i.e., promotion of enteral

nutrition, asepsis regulations and training of nurses) led to an immediate and stable reduction of

NMR and birth-weight specific survival of newborns with a BW<1500 grams, but not with a BW<1000

grams, primarily after reduced incidence and mortality of sepsis [7]. Introduction of nasal CPAP at

a NICU in Nicaragua reduced mortality amongst total newborns receiving ventilation assistance

(i.e. either mechanical ventilation or CPAP) [8]. Improvement (i.e., new equipment, refurbishment

and training of personnel) of a newborn unit to a Level III NICU at a teaching hospital in Ghana led

to significant reduction of mortality amongst newborns with a BW<2500 grams, mostly secondary

to significantly reduced incidence of perinatal asphyxia [9].

In these studies, training and expansion of personnel was a universal denominator for

improvement of care, which was also part of our intervention. Systematic training of midwives

in neonatal resuscitation has been a challenge in low resource countries and so far has yielded

positive results only in low risk settings, and takes time with need for strong re-enforcement

and repetition before an effect on neonatal mortality is observed [10-12]. However, increasing

the number of nurses per infant at the NICU may have a beneficial effect on neonatal outcome

[13,14]. Further improvement of survival may then be accomplished with increased capacity for

neonatal intensive care (e.g., increased capacity for (modernized) ventilation). We observed

a significant increase of use of neonatal intensive care commodities in the post-transition period.

Indeed, both higher level and volume of neonatal intensive care have been associated with

better survival of newborns with a BW<1500 grams [15,16]. While this seems an intuitive and logical

effect, it is important to realize that positive effects of higher capacity can only be sustained with

continuous and balanced availability of trained personnel, which can be challenging in the lower

resource setting [17,18]. Illustratively, in our population the reduction of admission rates in the post

transition period coinciding with increased number of nurses per bed may have been beneficial

for survival. However, the amount of nurses per infant at our facility is still less than recommended

for the intended level of care (i.e., one nurse per one or two beds), which may partially explain our

finding that the mortality rate in the most vulnerable small preterm infants (i.e., with a BW<1000

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

38

2

grams and <28 weeks of GA) did not decrease [19]. However, restricting the number of beds in

case of understaffing is extremely difficult when there are no other NICU level referral options

in Suriname.

Admission of more outborn neonates indicates an enhanced regional function of our neonatal

care facility, which was shown to be beneficial for their survival depending on the referral system. In

Ghana, survival of outborn newborns at the refurbished NICU was only beneficial to those referred

from private health facilities [9]. In our population, outborn newborns, mostly referred from birth

clinics and private or public Level II SNCUs at other hospitals, died more frequently than inborn

ones in both periods. Delays in transfer or higher prevalence of antenatal (e.g., preeclampsia) and

neonatal (e.g., prematurity) risk factors could have contributed to this [20-22]. However, the fact

that in our study demographics of outborn newborns were similar in both periods indicates

that better survival after the transition was mostly due to enhanced neonatal intensive care,

independent of presence of antenatal and neonatal risk factors. Screening regimens for antenatal

risk factors at surrounding birth clinics and in-utero transfer to our birth clinic, thereby creating

proximity to our neonatal care facility, could further enhance tertiary function and improve

survival in Suriname [23,24].

Mortality due to both perinatal asphyxia and sepsis were reduced in the post transition period.

For inborn newborns, training of obstetric nurses may have contributed to the reduction in

mortality of sepsis and similarly to less cases and better outcome of asphyxia. Additionally, for

both inborn and outborn newborns efficient treatment (e.g., modern equipment for mechanical

ventilation or circulatory support) at our refurbished NICU could have had beneficial effect on

survival of both. In the case of late-onset sepsis, incidence and mortality remained the same

after the transition. This indicates that our asepsis interventions, aimed primarily at prevention of

transmission of pathogens, failed, which is also reflected in similar amounts of ESBL-positive blood

cultures among both study periods. These results stress that in our setting strict enforcement of

asepsis protocol remains challenging, but should be prioritized.

Mortality of newborns with a BW<1000 grams remained high after the intervention. High

mortality of newborns with a BW<1000 grams was also observed in earlier reports in a Level II

SNCU in Jamaica, a Level III neonatal care facility in South Africa and at multiple NICUs in Brazil

and around the world [1,25-27]. In our low-resource setting, the fact that these newborns demand

a disproportionate share of scarcely available human and non-human recourses is a significant

limitation for improvement. However, almost half of them died of late-onset sepsis, indicating that

more effective infection prevention, including antibiotic stewardship, might substantially increase

their survival rates. Additionally, a major cause for morbidity amongst newborns with a BW<1500

grams in our study was NEC (Table 3b). Prevalence of NEC remained high, despite promotion

of feeding with human breast milk. Recent evidence from NICUs in developed countries has

shown that simple interventions (i.e., early human milk feedings, rigorous feeding protocol and

restricted feeding during indomethacin treatment and blood transfusions, and selective antibiotic

usage) can reduce incidence of NEC [28]. These interventions are cost-effective and can also easily

be applied in lower resource settings [29]. A major limitation in our setting is the unavailability

of total parenteral nutrition, but at the same time the low adherence to breast milk offers

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

39

2

a major opportunity for improvement. Lastly, the increased number of cases of NEC, along with

the increase in incidence of BPD, may be the unfortunate effect of more intensive care (e.g., more

ventilation, more early antibiotics) and better survival.

Limitations to this study were missing data (e.g., scarce data on additional outcomes

such as intraventricular hemorrhage, retinopathy of the premature, post-discharge survival),

the retrospective nature of this study, and relatively small numbers for complications with a low

incidence. Although we collected data to determine the highest level of care, we were not able to

apply an index to indicate severity of disease of newborns.

CONCLUSIONSThis study shows that scaling up of neonatal intensive care in Suriname substantially reduced

mortality of both in and outborn newborns through its enhanced availability and centralization.

Challenges ahead are sustainability, further improvement of tertiary function, and prevention of

sepsis and NEC with implementation of cost and resource effective interventions.

List of Abbreviations

SNCU = sick newborn care unit

NICU = neonatal intensive care unit

AHP = Academic Hospital Paramaribo

NMR = neonatal mortality rate (i.e., number of neonatal deaths per 1000 live births)

BW = birth weight

GA = gestational age

NEC = necrotizing enterocolitis

BPD = bronchopulmonary dysplasia

VAP = ventilator-associated pneumonia

EOS = early onset sepsis

LOS = late onset sepsis

ESBL = extended spectrum beta-lactamase

Maroon = descendant from Africans that escaped slavery and established independent societies

(e.g., term predominantly used in South America and on Caribbean Islands)

DECLARATIONSEthics

The Suriname Commission for Human Research approved this study (VG-021-14A).

Availability of Data and Materials

The datasets during and/or analyzed during the current study are available from the corresponding

author on reasonable request.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

40

2

Competing Interests

The authors declare that they have no competing interests.

Funding

R. Zonneveld was supported by the Thrasher Research Fund (TRF13064).

Authors’ Contributions

RZ, FBP and AJ conceived of the study. RZ, NH, FB, and AB performed the data search and

analysis. NPAL and PHD contributed in analyzing the data. All authors contributed to drafting

the manuscript, and read and approved the final manuscript.

Acknowledgments

The authors acknowledge the efforts of the employees of the medical archives of the Academic

Hospital Paramaribo for help with the retrieval of all patient charts used for this paper. We also

thank Professor Frans J. Walther at Leiden University and University of California Los Angeles for

careful review of this paper.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

41

2

REFERENCES1. Chow S, Chow R, Popovic M, Lam M, Popovic

M, Merrick J, Stashefsky Margalit RN, Lam H,

Milakovic M, Chow E, Popovic J. A Selected Review

of the Mortality Rates of Neonatal Intensive Care

Units. Front Public Health, 2015;7;3:225.

2. Carlo WA, Travers CP. Maternal and neonatal

mortality: Time to act J Pediatr (Rio J), 2016;426.

3. Black RE, Levin C, Walker N, Chou D, Liu L,

Temmerman M; DCP3 RMNCH Authors Group.

Reproductive, maternal, newborn, and child health:

key messages from Disease Control Priorities 3rd

Edition. Lancet 2016, S0140-6736(16)00738-8.

4. Knippenberg R, Lawn JE, Darmstadt GL,

Begkoyian G, Fogstad H, Walelign N, Paul

VK; Lancet Neonatal Survival Steering Team.

Systematic scaling up of neonatal care in

countries. Lancet 2005, 365(9464):1087-98.

5. Hedstrom A, Ryman T, Otai C, Nyonyintono

J, McAdams RM, Lester D, Batra M.

Demographics, clinical characteristics and

neonatal outcomes in a rural Ugandan NICU.

BMC Pregnancy Childbirth. 2014, 19;14:327.

6. Sen A, Mahalanabis D, Singh AK, Som TK,

Bandyopadhyay S. Impact of a district level sick

newborn care unit on neonatal mortality rate:

2-year follow-up. J Perinatol 2009, 29(2):150-5.

7. Agarwal R, Agarwal K, Acharya U, Christina

P, Sreenivas V, Seetaraman S. Impact of

simple interventions on neonatal mortality

in a low-resource teaching hospital in India. J

Perinatol 2007, 27(1):44-9.

8. Rezzonico R, Caccamo LM, Manfredini V,

Cartabia M, Sanchez N, Paredes Z, Froesch P,

Cavalli F, Bonati M. Impact of the systematic

introduction of low-cost bubble nasal CPAP in

a NICU of a developing country: a prospective

pre- and post-intervention study. BMC

Pediatr 2015, 25;15:26.

9. Enweronu-Laryea CC, Nkyekyer K, Rodrigues

OP. The impact of improved neonatal

intensive care facilities on referral pattern and

outcome at a teaching hospital in Ghana. J

Perinatol 2008, 28(8):561-5.

10. Carlo WA, Goudar SS, Jehan I, Chomba E,

Tshefu A, Garces A, Parida S, Althabe F, McClure

EM, Derman RJ, Goldenberg RL, Bose C, Krebs

NF, Panigrahi P, Buekens P, Chakraborty H,

Hartwell TD, Wright LL; First Breath Study

Group. Newborn-care training and perinatal

mortality in developing countries. N Engl J

Med 2010, 362(7):614-23.

11. Carlo WA, McClure EM, Chomba E,

Chakraborty H, Hartwell T, Harris H, Lincetto

O, Wright LL. Newborn care training of

midwives and neonatal and perinatal

mortality rates in a developing country.

Pediatrics 2010, 126(5):e1064-71.

12. Matendo R, Engmann C, Ditekemena J, Gado

J, Tshefu A, Kinoshita R, McClure EM, Moore

J, Wallace D, Carlo WA, Wright LL, Bose

C. Reduced perinatal mortality following

enhanced training of birth attendants in

the Democratic Republic of Congo: a time-

dependent effect. BMC Med 2011, 9:93.

13. American Academy of Pediatrics Committee

on Fetus and Newborn. Levels of neonatal

care. Pediatrics 2012, 130(3):587-97.

14. Callaghan LA, Cartwright DW, O’Rourke P,

Davies MW. Infant to staff ratios and risk of

mortality in very low birthweight infants. Arch

Dis Child Fetal Neonatal Ed 2003, 88(2):F94-7.

15. Phibbs CS, Baker LC, Caughey AB, Danielsen

B, Schmitt SK, Phibbs RH. Level and volume

of neonatal intensive care and mortality

in very-low-birth-weight infants. N Engl J

Med 2007, 356(21):2165-75.

16. Bartels DB, Wypij D, Wenzlaff P, Dammann

O, Poets CF. Hospital volume and neonatal

mortality among very low birth weight infants.

Pediatrics 2006, 117(6):2206-14.

17. Neogi SB, Malhotra S, Zodpey S, Mohan P.

Does facility based newborn care improve

neonatal outcomes? A review of evidence.

Indian Pediatr 2012, 49(8):651-8.

18. Neogi SB, Malhotra S, Zodpey S, Mohan

P. Challenges in scaling up of special care

newborn units--lessons from India. Indian

Pediatr 2011, 48(12):931-5.

19. British association of perinatal medicine.

Optimal arrangements for neonatal intensive

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

42

2

care units in the UK including guidance on their

medical staffing. A framework for practice. 3rd

edition 2014.

20. Sen A, Mahalanabis D, Singh AK, Som

TK, Bandyopadhyay S. Development and

effects of a neonatal care unit in rural India.

Lancet 2005, 366(9479):27-8.

21. Sehgal A, Roy MS, Dubey NK, Jyothi MC. Factors

contributing to outcome in newborns delivered

out of hospital and referred to a teaching

institution. Indian Pediatr 2001, 38(11):1289-94.

22. Arad I, Braunstein R, Bar-Oz B. Neonatal

outcome of inborn and outborn extremely

low birth weight infants: relevance of perinatal

factors. Isr Med Assoc J 2008, 10(6):457-61.

23. Chien LY, Whyte R, Aziz K, Thiessen P, Matthew

D, Lee SK: Canadian Neonatal Network.

Improved outcome of preterm infants when

delivered in tertiary care centers. Obstet

Gynecol 2001, 98(2):247-52.

24. Lorch SA, Baiocchi M, Ahlberg CE, Small DS.

The differential impact of delivery hospital

on the outcomes of premature infants.

Pediatrics 2012, 130(2):270-8.

25. Ballot DE, Chirwa TF, Cooper PA. Determinants

of survival in very low birth weight neonates in

a public sector hospital in Johannesburg. BMC

Pediatr 2010;10:30.

26. Trotman H. Bell Y. Neonatal sepsis in very

low birth weight infants at the University

Hospital of the West Indies. West Indian

Med J. 2006, 55:165-9.

27. Guinsburg R, de Almeida MF, de Castro JS,

Silveira RC, Caldas JP, Fiori HH, do Vale MS,

Abdallah VO, Cardoso LE, Alves Filho N,

Moreira ME, Acquesta AL, Ferrari LS, Bentlin

MR, Venzon PS, Gonçalves Ferri WA, Meneses

Jdo A, Diniz EM, Zanardi DM, Dos Santos

CN, Bandeira Duarte JL, Rego MA. Death

or survival with major morbidity in VLBW

infants born at Brazilian neonatal research

network centers. J Matern Fetal Neonatal

Med 2016, 29(6):1005-9.

28. Talavera MM, Bixler G, Cozzi C, Dail J, Miller RR,

McClead R Jr, Reber K. Quality Improvement

Initiative to Reduce the Necrotizing

Enterocolitis Rate in Premature Infants.

Pediatrics 2016, 137(5).

29. Johnson TJ, Patel AL, Bigger HR, Engstrom

JL, Meier PP. Cost savings of human milk

as a strategy to reduce the incidence of

necrotizing enterocolitis in very low birth

weight infants. Neonatology 2015, 107(4):271-6.

NEO

NA

TAL IN

TENSIV

E CA

RE IN SU

RINA

ME

43

2

SUPPLEMENTAL DATA

Supplemental Table 1. Local criteria for medium, high or intensive care

Criteria Medium care High care Intensive care

Gestational age (weeks) > 37 32-37 <32

Birth weight (grams) >20001 1000-2000 <1000

Respiratory None Nasal oxygen

cannula

Mechanical ventilation

CPAP

Circulatory None Peripheral vein

cannula

Arterial line

Cardiotonics

Gastro-intestinal Complete oral feeding Tube feeding Parenteral feeding

Gastro-intestinal surgery

Metabolic Phototherapy (at term)

Hypoglycemia

Phototherapy

Hematologic Blood transfusion Thrombocytes, FFP

Exchange transfusion

Neurologic Seizures

Perinatal asphyxia

Cerebral ultrasound

Infection Observation after

maternal risk factors2

Sepsis, meningitis, pneumonia

Other Congenital heart defects

CPAP = continuous positive airway pressure; FFP = Fresh Frozen Plasma. 1 At term dysmaturity was also a criteria for admission to the MC.2 Prolonged rupture of membranes (PROM), intrapartum fever and/or antibiotics, positive maternal Group-B

streptococcus culture.

II Prediction of Early Onset Sepsis

3 Association between Early Onset Sepsis Calculator and Infection Parameters for

Newborns with Suspected Early Onset Sepsis

Niek Achten, Rens Zonneveld, Ellen Tromp, Frans B. Plötz

J Clin Neonatol 2017, 6:159-62

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

52

3

ABSTRACTContext

Early onset sepsis (EOS) remains an important clinical problem with significant antibiotic

overtreatment as a result of poor specificity of clinical and infection parameters. Quantitative risk

stratification models such as the EOS calculator are promising, but it is unclear how these models

relate to infection parameters in the first 72 hours of life.

Aim

To evaluate the hypothesis that higher EOS calculator results are associated with (serial) laboratory

infection parameters, in particular an increase in CRP within 24-48 hours, and low leukocyte counts.

Design and Methods

EOS risk estimates were determined for infants born ≥ 34 weeks of gestation who were started

on antibiotic treatment for suspected EOS within 72 hours after birth. EOS risk estimates were

retrospectively compared to (changes in) available laboratory infection parameters including

C-reactive protein (CRP), leukocyte and thrombocyte count.

Statistical Analysis Used

Spearman’s rho rank correlations coefficient was used when testing for correlations between

continuous parameters. Kruskal-Wallis and Mann-Whitney U tests were applied to differences

between stratified risk groups.

Results

EOS risk was not correlated with changes in infection parameters. We found negative correlations

between both EOS risk and CRP level and leukocyte count within 6 hours of the start of antibiotics,

as well as CRP level between 6-24 hours after start of treatment.

Conclusions

In contrast to our hypothesis, high EOS risk at birth was consistently correlated with lower CRP and

leukocyte counts within 24 hours after the start of antibiotics, but not with infection parameters

after 24 hours. Further interpretation of infection parameters during sepsis calculator use needs

to be elucidated.

Key Messages

The sepsis calculator is neither associated with changes in CRP level, nor leukocyte or thrombocyte

count in newborns with suspected early onset sepsis.

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

53

3

INTRODUCTIONEarly onset neonatal sepsis (EOS) remains an important clinical problem in neonatal care. Due to

poor specificity of clinical findings and limited usability of available infection biomarkers, there is

significant over-treatment with antibiotics in the first 72 hours of life of newborns with suspected

EOS [1]. In an attempt to overcome this problem a quantitative risk stratification strategy based on

objective maternal risk factors and neonatal clinical findings has been developed [2]. This model,

hereafter referred to as the EOS calculator, provides a quantitative estimation of EOS risk along

with a recommendation on the use of antibiotics, and is available online. Two retrospective studies

revealed that application of the sepsis calculator may significantly reduce antibiotic therapy and

thus use of the EOS calculator may become more common in clinical practice [3,4].

Despite this promising potential, it is currently unclear how the EOS calculator estimated risk

and recommendations relate to infection parameters in the first 72 hours of life. Serial values in

C-reactive protein (CRP) and leukocyte count are still commonly used as arguments for the start

and duration of antibiotic therapy [1,5]. For this study, our aim was to evaluate the hypothesis that

higher EOS calculator results are associated with (serial) laboratory infection parameters. As EOS

is associated with elevated CRP and a lower leukocyte count [5,6] we particularly hypothesized

high EOS risk estimate to be associated with an increase in CRP within 24-48 hours, and low

leukocyte counts

SUBJECTS AND METHODSStudy Design

Data from a previously established retrospective birth cohort were used for analysis [4]. The study

included all newborns born ≥34 weeks of gestation, who were started on antibiotic treatment for

suspected EOS within 72 hours after birth, in Tergooi Hospital, Blaricum, The Netherlands, during

2014. Exclusion criteria were major congenital anomalies, including chromosomal, and prophylactic

treatment with antibiotics. The study was approved by the Scientific Review Committee of

Tergooi Hospital.

Data Collection

Maternal and neonatal clinical data were derived from hospital records. Local protocol required

routine infection parameter testing in newborns treated for clinically suspected EOS at start

of antibiotic therapy, and follow-up testing at 12-24 hours and/or 24-72 hours after the start of

antibiotic treatment. Infection parameter results were derived from electronic laboratory records.

EOS Calculator Risk Estimates and Stratification

EOS risk estimates were determined using the online calculator as provided by Escobar et al.,

through http://newbornsepsiscalculator.org [2,7]. These estimates represent the estimated

incidence of EOS per 1000 live births, and were calculated individually for each newborn in

the study. The resultant sepsis risk was categorized into three levels; <0.65 (low risk), 0.65-1.54

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

54

3

(intermediate risk), and >1.54 (high risk) per 1000 live births. In addition to using EOS calculator

risk estimate as continuous variables, we used these groups for stratified analysis.

Delta Variables

Since specifically serial values in infection parameters are used to guide clinical decisions [8] we

calculated delta variables when serial values were available. For delta variables, we calculated

absolute differences between values derived from initial blood draw (0-6 hours after start of

treatment) and follow-up values 24 hours after start of treatment. Values derived between 6-24

hours were used as follow-up values if values >24 hours were unavailable.

Statistical Analysis

All data were statistically analyzed using R (version 3.2.1) (http://www.r-project.org). Distributions

of continuous variables were visualized using kernel density plots. Spearman’s rho rank correlations

coefficient was used when testing for correlations between EOS risk estimates and infection

parameters (continuous variables not normally distributed). Kruskal-Wallis and Mann-Whitney U

tests were applied to determine significance of differences between EOS stratified risk groups.

RESULTSAfter exclusion of three newborns with insufficient clinical information to estimate EOS risk, data

from 108 newborns were used for analysis (Table 1).

CRP

We found negative correlations between EOS risk estimations and CRP levels within 6 hours and

between 6 and 24 hours after the start of antibiotics (Spearman’s rho -0.45 and -0.24, respectively).

This was confirmed by EOS stratified group analysis, where the high EOS risk group was associated

with lower CRP levels (<1 versus 11.5 mg/l, p<0.05, Table 1). EOS risk estimate was not correlated with

change in CRP as determined by the delta CRP variable based on serial CRP values.

Leukocytes and Thrombocytes

EOS risk estimate was not correlated with changes in serial leukocytes count. Lower leukocyte

counts within 6 hours after the start of antibiotics were associated with higher EOS risk estimations

(Spearman’s rho -0.30). Leukocyte count within 6 hours after start of antibiotics was lower in

the high-risk group compared to the intermediate/low risk group (p<0.05) (Table 1). There were

no correlations between EOS risk and (serial) thrombocyte counts.

DISCUSSIONIn contrast to our hypothesis, we did not find any correlations between EOS risk and changes in

serial CRP or serial leukocyte or trombocyte counts. We observed negative correlations between

EOS risk estimate and CRP level and leukocyte count within 6 hours of start of antibiotics, as well

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

55

3

as CRP level between 6-24 hours after start of treatment. Analyzing differences between EOS

stratified risk groups, comparable results within 6 hours of start of treatment were found.

In the high-risk group single point measurement CRP levels were in the normal range at start of

antibiotic therapy, which was started shortly after birth. This can be explained by the fact that CRP

levels represent endogenous neonatal synthesis, rise above 5 mg/l by 6-8 hours and peak around

24-48 hours [9,10]. Negative correlation between high EOS risk and CRP levels at the start of

antibiotic treatment may be explained by the fact that high-risk newborns started with antibiotic

treatment shortly after birth, before endogenous synthesis of CRP occurred. Furthermore, this

may also explain the significant differences of CRP levels of <1 mg/l in high-risk group versus

11.5 mg/l in the low-risk group at the start of antibiotics (P<0.05). In contrast to the high-risk

group, antibiotic therapy was mostly started 12 hours after birth in the low risk group of our

population [4].

Remarkably, CRP levels did not clearly increase in the high-risk group, as CPR level after

24-48h was not significantly raised compared to low and intermediate risk groups. This appears

to contrast with studies confirming that the sensitivity of CRP increases substantially with serial

determinations of CRP 24-48h after the onset of symptoms [9]. However, although EOS-risk is

correlated with infection, still the majority of the newborns in this group had negative blood

cultures, corresponding with persistent low CRP levels. In addition, given the half-life of CRP (19

hours) and clinical studies showing CRP levels decreasing after 16 hours in response to successful

antibiotic therapy, it is well possible that CRP-levels have returned to normal range within 24-48

Table 1. Infection parameters and correlation results among total and stratified risk group analysis.

EOS risk group

Overall

(n=108)

Stratified risk group analysis

Low

(n=41)

Intermediate

(n=10)

High

(n=57)

Infection parameter Median (IQR) n (%) Spearman’s rho Median (IQR)4

CRP

(median, mg/l)

<6 h <1 (13) 100 (93.6) -0.453 11.5(25) <1(8) <1(3)3

6-24 h 7 (23) 82 (75.9) -0.241 11.5(25) 2(8) 5(20)

>24 h 5.5 (17) 58 (53.7) -0.01 7(15) 3(20) 5(26)

Delta 4 (19) 96 (88.9) -0.08 6 (18) 2(21) 4(19)

Leukocytes

(median, x109/L)

<6 h 16.4 (9) 102 (94.4) -0.302 20.6(11) 15.3(20) 15.3(9)2

6-24 h 16.5 (10) 70 (64.8) -0.13 16.6(11) 26.2(15) 14.4(10)

>24 h 13.1 (7) 53 (49.1) -0.19 15.0(5) 14.3(16) 11.4(6)

Delta 3.7 (6) 85 (78.7) -0.18 4.7(9) 8.1(8) 2.9(5)

Trombocytes

(median, x109/L)

<6 h 219 (87) 94 (87.0) 0.04 224(112) 217(93) 215(92)

6-24 h 214 (113) 67 (62.0) 0.11 208(159) 233(91) 208(113)

>24 h 245 (106) 49 (45.4) 0.01 241(169) 280(80) 243(105)

Delta 27 (39) 77 (71.3) 0.09 25(57) 32(24) 27(39)

Statistically significant results marked in bold; 1P<0.05, 2P<0.01, 3P<0.001. 4 Mann-Whitney U test, high versus low/intermediate risk group. EOS risk; early onset sepsis risk as calculated with

sepsis calculator.

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

56

3

hours in infected children in the high-risk group, as this group was generally started on antibiotics

within shortly after birth [9,11].

From a clinical point of view these findings underline the puzzling nature of EOS clinical

management, with high EOS risk associated with low CRP levels. In the high-risk group, based

on objective maternal factors and newborn clinical evaluation, antibiotic therapy is started

and continued for 7 days. In this group, (serial) CRP measurement is not of additional value to

discontinue antibiotic therapy in case of negative blood cultures. In the low EOS risk group,

however, serial CRP may serve to discontinue antibiotic treatment after 3 days, given the negative

predictive value of serial low CRP levels [10].

The correlation between higher EOS risk estimates and lower leukocyte counts within 6 hours

after start of antibiotics corresponds with published findings showing lower leukocyte counts

being associated with EOS [6]. It should be noted however, that low leukocyte counts are rare

– reflected in a modest difference in absolute leukocyte count between the high-risk group and

overall median (15.3 vs 16.4 x109/L). Therefore, leukocyte counts are likely to be of limited clinical

value in EOS diagnostics. Finally, (changes) in thrombocyte counts were, in line with published

literature, not related to EOS risk. Thus we do not recommend the use of thrombocyte counts to

guide clinical decisions regarding antibiotics for EOS, regardless of estimated EOS risk.

Limitations of this study include its retrospective nature and selection bias for determination

of infection parameters. However, given the high percentage of available results within 6 hours of

start of antibiotics, we think this bias is limited for the correlations we found. Our sample size is

limited, but given the consistent results among correlation and stratified group level analysis, we

do not expect different results with a larger sample size.

In conclusion, EOS remains an important clinical problem with significant antibiotic

overtreatment as a result of poor clinical and infection parameters. In newborns treated for EOS,

risk estimates are neither associated with changes in CRP level, nor leukocyte or thrombocyte

count. If more widespread use of the sepsis calculator is expected, the interpretation of common

infection parameters in the context of EOS risk needs to be further elucidated.

EARLY

ON

SET SEPSIS CA

LCU

LATO

R

57

3

REFERENCES1. van Herk W, Stocker M, van Rossum AMC.

Recognising early onset neonatal sepsis: An

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77–82.

2. Escobar GJ, Puopolo KM, Wi S, Turk BJ,

Kuzniewicz MW, Walsh EM, Newman TB,

Zupancic J, Lieberman E, Draper D. Stratification

of risk of early-onset sepsis in newborns ≥ 34

weeks’ gestation. Pediatrics 2014, 133(1):30–6.

3. Shakib J, Buchi K, Smith E, Young PC.

Management of newborns born to mothers with

chorioamnionitis: Is it time for a kinder, gentler

approach? Acad Pediatr 2015, 15(3):340–4.

4. Kerste M, Corver J, Sonnevelt MC, van Brakel

M, van der Linden PD, M. Braams-Lisman BA,

Plötz FB. Application of sepsis calculator in

newborns with suspected infection. J Matern

Neonatal Med 2016, 7058:1–6.

5. Chirico G, Loda C. Laboratory aid to

the diagnosis and therapy of infection in

the neonate. Pediatr Rep 2011, 3(e1):1–5.

6. Newman TB, Puopolo KM, Wi S, Draper D,

Escobar GJ. Interpreting complete blood

counts soon after birth in newborns at risk for

sepsis. Pediatrics 2010, 126(5):903–9.

7. Puopolo KM, Draper D, Wi S, Newman TB,

Zupancic J, Lieberman E, Smith M, Escobar GJ.

Estimating the probability of neonatal early-

onset infection on the basis of maternal risk

factors. Pediatrics 2011, 128(5):e1155-63.

8. van Herk W, el Helou S, Janota J, Hagmann C,

Klingenberg C, Staub E, Giannoni E, Tissieres

P, Schlapbach LJ, van Rossum AM, Pilgrim SB,

Stocker M. Variation in Current Management

of Term and Late-Preterm Neonates at Risk for

Early-Onset Sepsis “An International Survey

and Review of Guidelines”. Pediatr Infect

Dis J 2016, 35(5):494–500.

9. Hofer N, Zacharias E, Müller W, Resch B. An

update on the use of C-reactive protein in

early-Onset neonatal sepsis: Current insights

and new tasks. Neonatology 2012, 102(1):25–36.

10. Simonsen KA, Anderson-Berry AL, Delair SF,

Dele Davies H. Early-onset neonatal sepsis.

Clin Microbiol Rev 2014, 27(1):21–47.

11. Ehl S, Gehring B, Pohlandt F. A detailed analysis

of changes in serum C-reactive protein levels

in neonates treated for bacterial infection. Eur

J Pediatr 1999, 158(3):238–42.

4 Immature-to-total-granulocyte Ratio as a Guide for Antibiotic Treatment in Suspected Early Onset Sepsis in

Surinamese Newborns

Rens Zonneveld, Sheldon Simson, John Codrington, Amadu Juliana, Frans B. Plötz

Submitted

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

60

4

ABSTRACTObjective

Measurement of immature granulocytes may be helpful in management of early onset sepsis

(EOS) in newborns in developing countries. We evaluate early negative prediction of automated

measurement of a one-point measurement of immature-to-total-granulocyte (I/T) ratio in

newborns with suspected EOS to help decisions on duration of antibiotic treatment.

Methods

A retrospective study was performed amongst newborns with a gestational age ≥34 weeks with

suspected EOS in whom local protocol had been followed for start and duration of antibiotic

treatment, and in whom granulocyte counts had been measured with a Sysmex XT 2000i

automated hematology analyzer.

Results

I/T and I/T2 were significantly lower (P 0.048 and P 0.015, respectively) in newborns with favorable

clinical course and in whom EOS was unlikely. In these newborns antibiotics were stopped 48-72

hours after start after which they all remained healthy.

Conclusion

Low I/T and I/T2 may help to increase the threshold to start empirical antibiotics or to guide safe

stoppage of antibiotics after 48-72 hours. Ultimately, this may help to reduce the antibiotic burden

in developing countries.

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

61

4

INTRODUCTIONAn estimated tenfold of newborns is empirically, and often unnecessarily, treated with antibiotics

in the first 72 hours of life for suspected early onset sepsis (EOS) [1]. Poor specificity of clinical

symptoms and limited utility of infection biomarkers complicate decisions on start and duration

of antibiotic treatment [1]. Over 60% of antibiotics given empirically before 72 hours of life are

prolonged beyond 48-72 hours despite a negative blood culture and a stable clinical condition

[2]. Furthermore, the threshold to start antibiotics in newborns in developing countries is low [3].

It remains a challenge to safely avoid unnecessary antibiotics in newborns with suspected EOS,

especially in developing countries.

Recent studies indicate that the immature-to-total-granulocyte (I/T) ratio may be promising

tool to identify infection at an early stage and to guide duration of antibiotic therapy (4-8).

Newman et al. proposed combination of the absolute neutrophil count (ANC) and I/T into the I/

T2, to be used in a prediction model for EOS [4]. Higher I/T2 predicted positive blood culture in

suspected EOS. Moreover, low serial immature-to-total-granulocyte ratios (I/T), in combination

with negative blood culture, can be used to discontinue antimicrobial therapy at 36-48 hours [5].

Whether a single point measurement of I/T or I/T2 at start of antibiotics in EOS is already predictive

for negative blood cultures is unknown.

Nowadays, the global availability of high-throughput flow-based automated hematology

analyzers (AHA) facilitates simple and fast measurement of immature granulocytes in developing

countries [6]. The aim of this study is to retrospectively evaluate early negative prediction

of automated measurement of I/T and I/T2 in Surinamese newborns with suspected EOS. We

hypothesized that I/T and I/T2 at start of antibiotic treatment were lower in newborns with unlikely

EOS in whom antibiotics were discontinued after 48-72 hours.

SUBJECTS AND METHODSStudy Design

A retrospective study was performed amongst newborns admitted to the neonatal care facility

at the Academic Hospital Paramaribo (AHP) from September 1st 2015 to May 30th 2016. Included

were newborns with a gestational age (GA) ≥34 weeks, who received antibiotic therapy within

72 hours after birth and with available laboratory results on immature granulocytes. Antibiotics

(intravenous ampicillin and gentamycin) were started for presence of maternal risk factors for EOS

or clinical suspicion. According to local protocol, EOS was considered unlikely if clinical course

had been uneventful (i.e., clinical improvement and no support other than normal volumes of IV

fluids or tube feeding), serial measurement of C-reactive Protein (CRP) was normal (i.e., levels <0.5

mg/dL), and blood culture was negative. Consequently, antibiotic treatment was discontinued

after 48-72 hours. Otherwise treatment was continued for 7 days. Newborns were divided in

two groups, namely: 1) Unlikely EOS: antibiotics discontinued after 48-72 hours; and 2) Probable

EOS: full 7-day treatment. The study protocol was part of a larger study on EOS, which was made

available on clinicaltrials.gov (NCT02486783) and was approved by the Surinamese Medical-Ethical

Board (VG-021-14A).

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

62

4

Data and Sample Collection

The following data were collected: maternal and prenatal conditions and risk factors, delivery

mode, GA, birth weight, ethnicity, gender, leukocyte differentials including immature granulocytes

and hour of life at blood draw, and hospital course. According to local protocol, determination

of leukocyte count was done in whole blood which was collected as part of standard draws for

infection parameters at t=0 (start of antibiotic treatment). In all newborns blood was collected for

bacterial culturing after insertion of a cannula before start of treatment.

Automated Measurement of Leukocytes

White blood cell (WBC) and platelet counts and ANC and IG (i.e., metamyelocytes, myelocytes and

promyelocytes) counts were collected from a Sysmex XT 2000i analyzer (Sysmex, Kobe, Japan). I/T

ratios were calculated as IG count divided by the total (ANC+IG) granulocyte count. We calculated

the I/T2 according to Newman et al. [7] by dividing the I/T by the ANC again.

Statistical Analysis

Categorical variables were presented as numbers and percentages and continuous variables

as mean with SD or, if not normally distributed, as median with IQR. Categorical variables were

compared with chi-square. Normality for continuous variables was assessed by a Shapiro-Wilk

test and further analyzed by a Student t-test or two-tailed Mann Whitney U-test. P <0.05 was

considered statistically significant.

RESULTSTwenty-seven newborns were included. Between both groups maternal, perinatal and neonatal

demographics were comparable (Table 1). In 13 (48.1%) newborns EOS was considered unlikely

and antibiotics were discontinued after 48-72 hours. Clinical course of these newborns had been

uneventful and they all remained healthy after stop of antibiotics. In the probable EOS group delta

CRP and need for additional support at t=48-72 was higher (Table 1). Of these newborns one had

a positive blood culture (i.e., Enterobacter cloacae) and one died after cardiorespiratory failure.

I/T and IT2 were significantly lower (P 0.048 and P 0.015, respectively) in newborns with

unlikely EOS versus probable EOS (Figure). When newborns under 4 hours of age were analyzed

separately, lower I/T2, but not lower I/T, was found in newborns with unlikely EOS (P 0.05 and

P 0.15 respectively).

DISCUSSIONIn this study, newborns with unlikely EOS had significantly lower I/T and I/T2 at start of antibiotic

treatment compared to newborns with probable EOS. The retrospective nature of this study

prevented bias in decision making for duration of antibiotic treatment based on AHA results. For

these reasons I/T and I/T2 may be highly useful in the clinic to guide antibiotic therapy in newborns

with suspected EOS in two ways. First, a low I/T and I/T2 may help to increase the threshold to start

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

63

4

Table 1. Descriptive statistics and laboratory results of Surinamese newborns with suspected early onset sepsis

(EOS).

Unlikely

EOS (N=13)

Probable

EOS (N=14) P value1

Maternal morbidity,

n (%)

Diabetes Mellitus

Hypertension

Preeclampsia

HIV

0

0

0

0

2 (14)

1 (7)

0

1 (7)

Mode of delivery,

n (%)

Vaginal

Caesarean

11 (85)

2 (15)

8 (57)

6 (43)

Male (female) 10 (3) 9 (5)

Ethnicity, n (%) Maroon or Creole

Other2

10 (77)

3 (23)

10 (71)

4 (29)

Gestational age,

median (weeks+days)

37+1 37+4

Birth weight,

mean±SD (grams)

2,628±245 2,731±152

Age at presentation3,

n (%)

<4 hours

≥4 hours

6 (46)

7 (54)

10 (71)

4 (29)

Clinical symptoms3,

n (%)

Respiratory distress

Circulatory instability

Feeding intolerance

Hypoglycemia

Lethargy

6 (46)

0

1 (8)

2 (15)

1 (8)

10 (71)

2 (14)

2 (14)

2 (14)

0

Reason for antibiotics,

n (%)

Prenatal risk factors

Clinical suspicion

6 (46)

7 (54)

4 (29)

10 (71)

Clinical Course4,

n (%)

CPAP

Mechanical Ventilation

Cardiotonics

Positive blood culture

Mortality

0

0

0

0

0

4 (29)

4 (29)

2 (14)

1 (7)

1 (7)

<0.001

CRP,

median (IQR) (mg/dL)

Initial3

Delta4

<0.5 (0)

0 (0.3)

<0.5 (2.8)

2.1 (3.7)

0.006

AHA measurements3,

median (IQR)

WBC (x109/L)

Platelets (x109/L)

ANC (x109/L)

IG%

16.4 (12.5)

219 (119)

11.7 (8.0)

0.7 (1.0)

17.0 (16.1)

193 (119)

7.9 (8.4)

1.2 (3.9)

AHA = automated hematology analyzer; HIV = human immunodeficiency virus; CPAP = continuous positive airway

pressure; CRP = C-reactive protein; WBC = white blood cell count; ANC = absolute neutrophil count; IG% = immature

granulocyte fraction.1 P >0.1 unless stated otherwise.2 Includes: Hindo-Surinamese, Javanese and Amerindian.3 At start of antibiotics.4 At t=48-72 hours after start of antibiotic treatment.

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

64

4 0.00

0.02

0.04

0.06

0.08

0.10

I/T

A

B

UnlikelyEOS (n=13)

Probable EOS (n=14)

0.000

0.002

0.004

0.006

0.008I/T

2

*

*

Figure 1. Immature-to-total-granulocyte ratios (I/T and I/T2) in Surinamese newborns treated with

antibiotics for suspected early onset sepsis (EOS). A: I/T; B: I/T2. Data represent ratios calculated from

results of automated measurement of granulocytes with a Sysmex XT 2000i analyzer in whole blood obtained

from newborns with suspected EOS at start of antibiotic treatment. I/T2 is calculated according to Newman et

al. (7). * P <0.05 compared to probable EOS. Bars represent median values and error bars interquartile range.

empirical antibiotic therapy. Second, it may guide clinicians to shorten antibiotic treatment to

48-72 hours in case of negative blood cultures.

Of the measured parameters, I/T2 showed the most significant difference between short and

full treatment duration. This is in line with the conclusions made by Newman et al. [4,8]. They

postulated that measurement of both the ANC and I/T have separate predictive values but are

two dependent tests since the ANC is a variable in the denominator of I/T. The I/T2 was designed

to capture both tests into one ratio, which showed increasing performance in newborns older

than 4 hours of life and when there was high suspicion of EOS. In contrast to Newman et al., we

were particularly interested in the negative predictive ability of I/T2 during the transition period

of newborns. In our cohort most newborns started within 4 hours of life with antibiotics. In these

newborns the threshold to start empirical antibiotic therapy is lower due to ambivalent clinical

symptoms during transition to extrauterine life. When measured within 4 hours after birth, we

observed a trend in lower I/T2 in cases of unlikely EOS. This is a clinically relevant finding, which

may favor the I/T2 in increasing the threshold to start antibiotics during the transition period in

newborns with a low suspicion of EOS.

Nowadays, CRP is frequently used in clinical settings to determine safe stoppage of antibiotics

for EOS, also in Suriname. Serially measured low levels of CRP predict absence of EOS with a negative

predictive value (NPV) of 99%, yet only in combination with a negative blood culture and clinically

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

65

4

improved newborn [2,9]. However, in practice, despite this high NPV, serial measurement of CRP

can also lead to additional testing and longer duration of antibiotic treatment [10]. Based on our

results, the NPV of a one-point measurement of I/T2 may be comparable with serial measurement

of CRP and should now be further evaluated in prospective studies. The I/T2 could then also be

added to an algorithm with serial measurement CRP in order to enhance NPV and clinical utility

of both markers [11]. Last, automated measurement of immature granulocytes performed at

least equal to manual differentiation [12]. The obvious advantages of automated measurement

are the large sample size, the immediate availability of test results for the clinician, and its cost-

effectiveness for the low-resource setting.

Because measurement of immature granulocytes was not part of standard care in Suriname,

the sample size for this study was relatively small. Larger and prospectively included cohorts are

necessary to increase prevalence of positive blood cultures and further controlling for maternal

(e.g., preeclampsia, mode of delivery) and neonatal (e.g., sex, ethnicity, hours of life) factors that

may influence leukocyte differentials [8,13]. However, we studied a homogeneous group of near

term and term newborns so we do not expect our results to differ in larger cohorts. Additionally,

establishing larger cohorts with blood culture confirmed EOS is complicated since prevalence of

positive bacterial blood cultures is low [1].

CONCLUSIONOur results indicate clinical utility of automated measurement of I/T and I/T2 in clinical decision-

making on start and duration of antibiotic treatment in EOS. Ultimately this may contribute to

reduction of the antibiotic burden in Suriname and other developing countries.

Abbreviations and Definitions

ANC = absolute neutrophil count

IG = immature granulocyte

IG% = immature granulocyte fraction of total leukocytes

I/T = immature-to-total-granulocyte ratio

I/T2 = immature-to-total-granulocyte ratio divided by the absolute neutrophil count

Maroon = descendant from Africans that escaped slavery and established independent societies

(e.g., term predominantly used in South America and on Caribbean Islands)

Acknowledgments

RZ was supported by a stipend from the Thrasher Research Fund (TRF13064) and from Tergooi

Hospitals, Blaricum, The Netherlands. The authors acknowled the efforts of all employees of

the Clinical Laboratory of the Academic Hospital Paramaribo for assistance with sample handling.

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

66

4

Author Contributions

R Zonneveld and FB Plötz designed the study. R Zonneveld, S Simson, A Juliana and J Codrington

collected and analyzed the data. R Zonneveld and FB Plötz drafted the manuscript. S Simson,

A Juliana and J Codrington revised and helped draft the final manuscript. All authors have read

and approved the final manuscript. The authors declare they have no conflict of interest.

IMM

ATU

RE GR

AN

ULO

CY

TES IN SU

RINA

MESE N

EWBO

RNS

67

4

REFERENCES

1. van Herk W, Stocker M, van Rossum AM.

Recognising early onset neonatal sepsis: an

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77-82.

2. Canty JB, Wozniak PS, Sanches PJ. Prospective

surveillance of antibiotic use in the neonatal

intensive care unit: results from the SCOUT

study. Pediatr Infect Dis J 2015, 34:267-72.

3. Hariharan S, Chen D, Harry C, Ragobar R,

Boodoosingh R, Gangoo C et al. Antimicrobial

prescription and usage in the neonatal

intensive care units of a Caribbean country:

a prospective observational study. J Neonatal

Perinatal Med 2013, 6(4):325-31.

4. Newman TB, Draper D, Puopolo KM, Wi S,

Escobar GJ. Combining immature and total

neutrophil counts to predict early onset sepsis

in term and late preterm newborns: Use of

the I/T2. Pediatr Infect Dis J 2014, 33:798–802.

5. Mikhael M, Brown LS, Rosenfeld CR. Serial

neutrophil values facilitate predicting

the absence of neonatal early-onset sepsis. J

Pediatr 2014, 164(3):522-8.

6. Zonneveld R, Molema G, Plötz FB. Analyzing

Neutrophil Morphology, Mechanics, and Motility in

Sepsis: Options and Challenges for Novel Bedside

Technologies. Crit Care Med 2016, 44(1):218-28.

7. Murphy K, Weiner J. Use of leukocyte counts

in evaluation of early-onset neonatal sepsis.

Pediatr Infect Dis J 2012, 31(1):16-9.

8. Newman TB, Puopolo KM, Wi S, Draper D,

Escobar GJ. Interpreting complete blood

counts soon after birth in newborns at risk for

sepsis. Pediatrics 2010, 126(5):903-909.

9. Hofer N, Zacharias E, Müller W, Resch B. An

update on the use of C-reactive protein in

early-Onset neonatal sepsis: Current insights

and new tasks. Neonatology 2012, 102:25–36.

10. Mukherjee A, Davidson L, Anguvaa L, Duffy

DA, Kennea N. NICE neonatal early onset

sepsis guidance: greater consistency, but more

investigations, and greater length of stay. Arch

Dis Child Fetal Neonatal Ed 2015, 100(3):F248-9.

11. Kerste M, Corver J, Sonnevelt MC, van Brakel

M, van der Linden PD, M Braams-Lisman BA et

al. Application of sepsis calculator in newborns

with suspected infection. J Matern Fetal

Neonatal Med 2016, 7:1-6.

12. MacQueen BC, Christensen RD, Yoder

BA, Henry E, Baer VL, Bennett ST et

al. Comparing automated vs manual

leukocyte differential counts for quantifying

the ‘left shift’ in the blood of neonates. J

Perinatol 2016, 36(10):843-8.

13. Wiland EL, Sandhaus LM, Georgievskaya Z,

Hoyen CM, O’Riordan MA, Nock ML. Adult

and child automated immature granulocyte

norms are inappropriate for evaluating

early-onset sepsis in newborns. Acta

Paediatr 2014, 103(5):494-7.

III The Vascular Pathophysiology of Early Onset Sepsis

5 Soluble Adhesion Molecules as Markers for Sepsis and the Potential

Pathophysiological Discrepancy in Neonates, Children and Adults

Rens Zonneveld, Roberta Martinelli, Nathan Shapiro, Taco W. Kuijpers, Frans B. Plötz, Christopher V. Carman

Critical Care 2014, 18:204

Editorial Comment in: McCulloh RJ, Spertus JA. Separating signal from noise: the challenge of

identifying useful biomarkers in sepsis. Crit Care 2014, 18(2):121

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

76

5

ABSTRACTSepsis is a severe and life-threatening systemic inflammatory response to infection that affects

all populations and age groups. The pathophysiology of sepsis is associated with aberrant

interaction between leukocytes and the vascular endothelium. As inflammation progresses,

the adhesion molecules that mediate these interactions become shed from cell surfaces and

accumulate in the blood as soluble isoforms that are being explored as potential prognostic

disease biomarkers. We critically review the studies that have tested the predictive value of soluble

adhesion molecules in sepsis pathophysiology with emphasis on age, as well as the underlying

mechanisms and potential roles for inflammatory shedding. Five soluble adhesion molecules are

associated with sepsis, specifically, E-selectin, L-selectin and P selectin, intercellular adhesion

molecule-1 and vascular cell adhesion molecule-1. While increased levels of these soluble adhesion

molecules generally correlate well with the presence of sepsis, their degree of elevation is still

poorly predictive of sepsis severity scores, outcome and mortality. Separate analyses of neonates,

children and adults demonstrate significant age-dependent discrepancies in both basal and

septic levels of circulating soluble adhesion molecules. Additionally, a range of both clinical

and experimental studies suggests protective roles for adhesion molecule shedding that raise

important questions about whether these should positively or negatively correlate with mortality.

In conclusion, while predictive properties of soluble adhesion molecules have been researched

intensively, their levels are still poorly predictive of sepsis outcome and mortality. We propose

two novel directions for improving clinical utility of soluble adhesion molecules: the combined

simultaneous analysis of levels of adhesion molecules and their sheddases; and taking age-related

discrepancies into account. Further attention to these issues may provide better understanding of

sepsis pathophysiology and increase the usefulness of soluble adhesion molecules as diagnostic

and predictive biomarkers.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

77

5

INTRODUCTIONSepsis [1], due to its detrimental sequelae and limited therapeutic options, continues to be

responsible for many deaths amongst all age groups [2-4]. Growing evidence indicates that

aberrant leukocyte activation and recruitment into host tissues plays a pivotal role in causing

breakdown of the vascular endothelium [5], which in turn leads to organ failure and death [6].

Inflammatory leukocyte recruitment is initiated by soluble mediators (for example, cytokines

or bacterial-derived lipopolysaccharide (endotoxin)), which upregulate adhesion molecule

expression on both leukocytes and the endothelium. This upregulation results in a multistep

adhesion cascade whereby circulating immune cells sequentially roll on, firmly adhere to, and

transmigrate across the endothelium [7-9]. During the progression of inflammatory responses,

soluble isoforms of the leukocyte recruitment adhesion molecules are shed from cell surfaces and

accumulate within the circulating blood plasma [10]. These soluble isoforms have been considered

promising prognostic biomarkers of severity of inflammation but the clinical utility of monitoring

such changes remains poor [11].

One reason for the thus far limited clinical utility of these soluble isoforms is the fact that

shedding in general is neither a passive nor an inevitable consequence of upregulated expression/

cell activation. Most shedding is an active process, which is discretely regulated by diverse

proteolytic enzymes, although cell damage can also variably contribute to soluble adhesion

molecule levels [10]. Although still a matter of controversy, there is increasing evidence

that shedding serves regulatory roles to dampen inflammation (and specifically to reduce

leukocyte– endothelial interactions) and protect the host from excessive collateral damage

[10,12]. Furthermore, age-related differences in both levels of soluble adhesion molecules and

the enzymes that mediate shedding have been observed in both healthy and septic patients (as

discussed in detail below). The relationship between soluble adhesion molecule levels, underlying

inflammatory and shedding activities and clinical outcomes may thus be more complex than

once thought.

The goals of this review are therefore to summarize existing knowledge regarding

the mechanisms and putative functions for shedding of cell surface adhesion molecules/

generation of soluble isoforms, unequivocally identified to exist at elevated levels in the blood of

septic patients, and to investigate how these levels and their shedding differ amongst healthy and

septic neonates, children and adults to improve our understanding and clinical utility of soluble

adhesion molecules.

LITERATURE SEARCHWe performed a comprehensive literature search in MEDLINE using medical subject headings

and text words, supplemented by scanning the bibliographies of the recovered articles. We

combined ‘endothelium’ and ‘leukocytes’ using the term ‘OR’. This search was subsequently

combined with ‘sepsis’ using the Boolean operator ‘AND’. We used a similar search strategy, using

the terms ‘soluble’ and ‘circulating adhesion molecules’. We combined these results with the terms

‘sepsis’, ‘septic shock’, ‘endothelium’, ‘leukocytes’, ‘monocytes’, ‘granulocytes’, ‘macrophages’,

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

78

5

‘neutrophils’, ‘lymphocytes’ and ‘inflammation’. We then combined these results with the terms

‘children’, ‘neonates’, ‘adults’ and ‘age’.

SOLUBLE ADHESION MOLECULES: FROM CELL SURFACE TO CIRCULATIONFive soluble adhesion molecules were associated with sepsis and their main characteristics are

summarized in Table 1. Three adhesion molecules (E-selectin, L-selectin and P-selectin) belong

to the selectin superfamily and function in leukocyte rolling (Figure 1). Two adhesion molecules

(intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1))

belong to the immunoglobin domain superfamily cell adhesion molecules that are important

for firm adhesion and transendothelial migration [13]. In all cases, inflammatory mediators (for

example, cytokines, thrombin, lipopolysaccharide) first increase cell surface expression of these

molecules followed by the later appearance of shed, soluble isoforms (Table 1 and Figure 2).

E-selectin

Soluble isoforms of E-selectin can be found in the supernatant of endothelial cells cultured in vitro

within 24 hours of cytokine activation and are generated through a largely caspase dependent

shedding process [10,14-16]. In healthy individuals low levels of soluble E-selectin (sE-selectin)

are found in serum, but these levels are greatly elevated in septic patients [16,17]. Importantly,

shed sE-selectin from sera of septic patients retained the ability to adhere to granulocytes in vitro

Table 1. Characteristics of adhesion molecules involved in sepsis

Adhesion

molecule Expression Ligands

Inflammatory

mediators

Mode of

expression

Specific

Function Sheddase

E-selectin Endothelial

cells

ESLG-1

PSGL-1

TNF-α, LPS,

IL-1

Inducible Rolling Caspase

L-selectin Leukocytes GlyCAM-1

MAdCAM-1

TNF-α, LPS,

IL-1, IL-6,

Constitutive

Inducible

Rolling ADAM-17

P-selectin Endothelial

cells

Platelets

PSGL-1 TNF-α, IL-4,

IL-13, histamine,

thrombin

Constitutive Rolling MMP

ICAM-1 Endothelial

cells

Mac-1,

LFA-1

TNF-α, LPS,

IL-1

Constitutive

Inducible

Firm adhesion

TEM

ADAM- 17

NE

VCAM-1 Endothelial

cells

VLA-4 TNF-α, LPS,

IL-1

Constitutive

Inducible

Firm adhesion

TEM

ADAM- 17

NE

ICAM-1 = intercellular adhesion molecule-1; VCAM-1 = vascular cellular adhesion molecule-1; ESGL-1 = endothelial selectin

glycoprotein ligand; PSGL-1 = platelet selectin glycoprotein ligand; GlyCAM-1 = glycosylation dependent cell adhesion

molecule; MAdCAM-1 = mucosal vascular addressin cell adhesion molecule; Mac-1 = macrophage antigen; LFA-1 = leukocyte

function antigen; VLA-4 = very late antigen; TNF- α =tumor necrosis factor alpha; LPS = lipopolysaccharide; IL = interleukin;

TEM = trans endothelial migration; ADAM-17 = A Disintegrin And Metalloproteinase; MMP = matrix metalloproteinase;

NE = neutrophil elastase.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

79

5

A Extravasation

C Trans-Cellular DiapedesisB Para-Cellular Diapedesis

Vascular lumen

Activation& Adhesion

Lateral MigrationRolling

Shear

Migration between endothelial cells

Migration though pore in an individual endothelial cell

Open inter-cellular gap

Diapedesis

* transmigratory cup

* * * *

VLA-4 Mac-1

(VCAM-1 / ICAM-1)

* transmigratory cup

(VCAM-1 / ICAM-1)

E-L-P-SelectinLFA-1

VCAM-1 ICAM-1

Open trans-cellular pore

Figure 1. A: Stages of extravasation of a leukocyte. Leukocytes first undergo tethering and rolling on

the endothelium, mediated by E-, L-, and P-selectins and their carbohydrate ligands; activation and adhesion:

leukocyte rolling facilitates interaction with chemoaatractants present on endothelial surfaces, which in turn

causes leukocyte activation that triggers firm adhesion and arrest, mediated by the integrins Mac-1, LFA-1 and

VLA-4 binding to their endothelial ligands ICAM-1 and VCAM-1. Subsequently, leukocytes engage in lateral

migration over the endothelial wall in search for a site to transmigrate, guided by VCAM-1/ICAM-1 enriched

transmigratory cups (asterisks in B and C), present on endothelial cells. The last step in this cascade is

transendothelial migration (TEM) or diapedesis, whereby the leukocytes cross the endothelial barrier, either

B: paracellular, through the inter-endothelial junctions, or C: transcellular, via the formation of a transcellular

pore. See reference 7-9 and 13 for additional details.

B Mechanisms for reduced adhesion after shedding

A Adhesion cascadeVLA-4

VCAM-1

(For simpli�cationonly illustrating thisreceptor-ligand pair)Rolling Activation & Adhesion

1. Decreased Cell Surface Density 2. Decoy Ligands

Lateral migration & Diapedesis

Sheddase

Figure 2. Shedding of selectin and immunoglobin superfamily adhesion molecules and its functional

implications. For simplification only one example receptor ligand pair (VLA-4/VCAM-1) is illustrated. A:

Interactions of cell surface integrin VLA-4, present on leukocytes, with endothelial cell surface VCAM-1 during

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

80

5[16]. Shedding of E-selectin has thus been proposed to limit leukocyte–endothelial interactions

both by decreasing the cell surface density on the endothelium and by generating an intravascular

competitive inhibitor or decoy ligand (that is, sE-selectin) for leukocytes, thereby reducing

collateral damage in the host [18]. Indeed, one clinical study found that while sE-selectin was

elevated in septic children, those with the highest levels exhibited the best outcomes and survival

rates [19].

L-selectin

Within approximately 10 to 15 minutes of leukocyte activation by cytokines (for example, tumor

necrosis factor alpha) or lipopolysaccharide, soluble L-selectin (sL-selectin) is measurable in

the blood plasma as a result of cleavage by a disintegrin and metalloproteinase (ADAM)-17

[10,20,21]. Clinical studies show that L-selectin is shed and detected at elevated levels in the plasma

during the systemic inflammatory response [22]. Interestingly, Seidelin and colleagues [22] found

that the greatest survival was among septic adults that presented with the highest levels of sL-

selectin, and similar findings were made in another study focused on children [19]. Additionally,

in an in vitro fluid shear flow model, exogenously added sL-selectin inhibited leukocyte rolling

and firm adhesion to the endothelium in a dose dependent manner, presumably by competing

with cell surface L-selectin for binding of endothelial ligands [23]. Moreover, Ferri and colleagues

have demonstrated that systemic administration of exogenous sL-selectin to mice in vivo reduced

intravascular leukocyte rolling and adhesion, and as a consequence decreased vascular leak in

models of both local inflammation and sepsis [24-26]. Alternatively, addition of a small molecule

inhibitor of shedding increased leukocyte adhesion and vascular leak in the same settings [26].

The authors thus propose a significant protective role for L-selectin shedding in sepsis.

P-selectin

P-selectin is found within both endothelial cells and platelets [27,28]. As with the other selectins,

P-selectin can be measured in its soluble form in cell culture supernatants and in blood plasma,

with greater levels found in septic patient plasma [29]. Mechanisms for P-selectin shedding remain

poorly characterized, although some experimental data show that shedding of P-selectin might

occur through cleavage by matrix metalloproteinase in patients with cardiovascular disease or

hypertension [30,31]. The degree to which plasma soluble P-selectin (sP-selectin) is derived from

endothelial cells versus platelets remains unclear. However, one study found a strong positive

different steps in the leukocyte adhesion cascade; B: Ectodomain shedding of cell surface VCAM-1 (thereby

generating the soluble isoform sVCAM-1) by sheddases might reduce leukocyte adhesion to the endothelium

on two complementary levels: 1) reduction of cell surface density of adhesion molecules (e.g., decreased

endothelial cell surface VCAM-1, in this example); 2) binding of the resulting soluble isoforms to cell surface

ligands (i.e., binding of sVCAM-1 to leukocyte cell surface VLA-4, in this example), thereby serving as

competitive antagonists (or decoy ligands) for the remaining cell-surface adhesion receptors. See reference

11 and the text for more details on functional implications of shedding.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

81

5

correlation between coagulation (disseminated intravascular coagulation, fibrinogen consumption

and thrombin activation markers) and sP-selectin in septic patients, indicating a significant role

for platelet shedding of P-selectin [32]. Independently of its sources, sP-selectin could negatively

modulate direct leukocyte-endothelial interactions and/or indirect platelet mediated secondary

capture of leukocytes on the endothelium (both of which are dependent on P-selectin– platelet

selectin glycoprotein ligand-1 adhesion), although this remains to be tested directly [28].

Intercellular Adhesion Molecule-1

In vitro 1 to 6 hours after activation of endothelial cells by cytokines, soluble ICAM-1 (sICAM-1) can

be measured in culture supernatants, after shedding mediated by neutrophil elastase-dependent,

ADAM-17-dependent and matrix metalloproteinase-9-dependent shedding [10,33-35]. Shedding

of ICAM-1 is thought to promote detachment of leukocytes from the endothelium, thus limiting

local inflammation [10]. Indeed, addition of exogenous neutrophil elastase was shown to be

highly effective at cleaving surface-bound human ICAM-1 in vitro, which in turn abrogated Mac-

1-dependent leukocyte adhesion [36]. Evidence supports a potentially protective role for ICAM-1

shedding during sepsis. Elevated levels of sICAM-1 are well documented in human sepsis [5,6,11],

and studies performed in both humans and mice demonstrate that this is induced within ~

4 hours of endotoxin challenge [37-39]. Significantly, septic children with the highest levels of

sICAM-1 had better outcomes/survival rates [19], suggesting that shedding and loss of cell surface

ICAM-1 from the endothelium may serve a protective function. Interestingly, in a cecal ligation

puncture model of sepsis, ICAM-1-knockout mice – whereby cell-surface ICAM-1 is completely

abolished – exhibited a significant decrease in leukocyte tissue invasion, organ damage and

mortality compared with wild-type mice [37].

Vascular Cell Adhesion Molecule-1

In vitro 1 to 6 hours after cytokine activation, VCAM-1 is measurable in its soluble form (sVCAM-1) in

endothelial cell supernatant through shedding mediated by ADAM-17, cathepsin G and neutrophil

elastase [10]. Singh and colleagues demonstrate that this may be achieved, at least in part,

through the cytokine-mediated (that is, interleukin-1β -mediated and tumor necrosis factor alpha-

mediated) downregulation of tissue inhibitor metalloproteinase-3, which they showed to function

as a tonic suppressor of ADAM-17-mediated VCAM-1 shedding [40]. In response to endotoxin,

sVCAM-1 was observed to be markedly elevated in mice [38] and humans [39]. Furthermore,

increased levels of sVCAM-1 are reported in human sepsis, and higher levels seem to be associated

with increased severity of disease and non-survival [5,6]. Shedding of VCAM-1 is implicated to

counteract the pro-adhesive state of leukocytes to the endothelium both by lowering endothelial

receptor density [10] and by forming sVCAM-1 to act as a competitive inhibitor (or decoy receptor)

of leukocyte very late antigen-4 [40,41] (Figure 2). Interestingly, prednisolone – a synthetic

glucocorticoid shown to be beneficial in the treatment of sepsis – was shown to enhance sVCAM-1

levels, suggesting the intriguing possibility that its mechanism of action may be at least partially

related to potentiation of VCAM-1 shedding [39].

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

82

5

LEVELS OF SOLUBLE ADHESION MOLECULES: IMPACT OF AGE Neonates

Basal levels of all soluble adhesion molecules of healthy neonates are comparable with (sICAM-1) or

higher than (sE-selectin, sP-selectin and sVCAM-1) basal levels in children or adults (Tables 2, 3, 4, 5

and 6 and Figure 3). Only the levels of sL-selectin are lower in neonates than in children or adults.

Importantly, in neonatal sepsis, levels of all soluble molecules are increased, but both the relative

and absolute extent of increase is remarkably lower compared with adults or children (Tables 2, 3,

4, 5 and 6 and Figure 3). This raises the important question of whether neonates are less effective

at shedding or less avidly upregulate adhesion molecules in the first place. Indeed, some studies

have been conducted to directly address this issue, which suggests contributions from both of

these mechanisms. Cell surface levels of L-selectin on neutrophils and sL-selectin levels from

cord blood were both lower than the cell surface and the circulating form of L-selectin found in

adult blood [42-44]. Interestingly, when challenged with neutrophil-activating chemoattractants,

neonatal neutrophils exhibited a significantly lower shedding response compared with adult

neutrophils [42,43]. Austgulen and colleagues suggest that these differences may be a reflection of

a developing immune system [45] that shows features of hyporesponsiveness (46). Since neonatal

sepsis/infection is particularly difficult to diagnose and no dependable predictors exist, sE-selectin

[45,47,48], sL-selectin [49,50] and sP-selectin [48,49] as well as sICAM-1 [50-59] and sVCAM-1 [49]

were evaluated as markers for the presence of infection in neonates. However, none of these

soluble isoforms were introduced in a clinical setting because they did not reach predictive ability.

The above discussion (and additional elaborations below) points toward complexities that need to

be resolved before meaningful interpretations can be made.

Children

Generally speaking, the basal levels of soluble adhesion molecules in healthy children are similar to

or lower than those of adults and neonates. However, both the relative amount and the absolute

amount of sE-selectin and sL-selectin during sepsis seem much higher, whereas sP-selectin

levels remain low. On the other hand, sICAM-1 and sVCAM-1 have similar basal levels and sepsis

generates comparable or higher levels versus adults (Tables 2, 3, 4, 5 and 6 and Figure 3). Three

studies assessed age-dependent differences in levels of selectins in healthy children [59,60,64].

Interestingly, infant had significantly lower levels of sL-selectin when compared with toddlers

(average age 2 years) [59]. Additionally, healthy children (age 9 to 15.5 years) were found to have

significantly decreasing sE-selectin levels with increasing age [64]. The authors of all three studies

suggest that potential developmental changes exist in both expression and shedding of selectins,

but that the physiological relevance of these observations remains to be determined.

During sepsis in children, studies show a significant increase in levels of soluble adhesion

molecules [19,61-63]. Interestingly, Briassoulis and colleagues show significant increase of sE-

selectin, as well as sL-selectin and ICAM-1, especially amongst survivors [19]. The authors conclude

that inadequate or suppressed shedding during sepsis might be associated with increased

mortality, and they hypothesize that the shedding process is indeed protective for the host.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

83

5

Tab

le 2

. Lev

els

of s

olu

ble

E-se

lect

in in

neo

nate

s, c

hild

ren

and

adul

ts

sE-s

elec

tin

Stud

y [r

efer

ence

]M

ean

ag

eSe

psi

s cr

iter

ia

Hea

lth

y

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

hea

lth

y)

Sep

sis

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

sep

sis)

Neo

nate

sD

olln

er e

t al

[52]

0-7

day

sC

linic

al91

.4 (

2-21

7.8)

2415

1.7

(37-

362.

2)18

Aus

tgul

en e

t al

[45]

Pre/

at te

rmC

linic

al13

4.1 (

69.5

-280

)16

818

7.7

(118

.4-2

62.2

)24

Edga

r et

al [

57]

0-7

day

sC

linic

al71

(51

-118

)27

135

(94-

192)

192

Gia

nnak

i et

al [4

7]A

t te

rmC

linic

al13

9 ±4

840

Edga

r et

al [

55]

At

term

Clin

ical

71 (

51-1

18)

4615

8 (9

4-20

7)46

Chi

ldre

nA

ndry

s et

al [

60]

6-10

57.6

(36

.7-1

52.2

)68

11-1

542

.1 (2

9.9-

114.

1)90

Paiz

e et

al [

61]

2-16

PRIS

M10

0 (

90-1

10)

4023

0 (

100

-380

)20

Kru

eger

et

al [6

2]3.

5 (0

.2-1

6)A

CC

P/SC

CM

68 (4

9-10

5)22

131 (

112-

146)

22

Wha

len

et a

l [63

]1d

to 17

yD

oug

hty

et a

l.246

±6

1423

077

Nas

h et

al [

64]

970

(35

-121

)81

1559

(25

-119

)

Bria

sso

ulis

et

al [1

9]6.

5 (2

.8)

PRIS

M16

1 ±43

1093

6 ±3

9910

Ad

ults

Pres

terl

et

al [6

5]51

(30

-67)

APA

CH

E II

48.9

(14

.3-8

9.9)

2013

0 (4

0-5

70)

20

Wei

gand

et

al [6

6]58

.7 (4

.4)

AC

CP/

SCC

M29

(14

.3-8

9.9)

785

14

Hyn

nine

n et

al [

67]

49 (

17.2

)A

PAC

HE

II73

(62

-89)

11

Kna

pp e

t al

[68]

51 (

21-9

6)A

PAC

HE

III43

.7 ±

20.3

1594

.5 ±

5428

Osm

ano

vic

et a

l [69

]A

dul

t28

.5 ±

14.3

1811

8 ±8

49

Sod

erq

uist

et

al [

70]

71 (

10-9

1)U

nkno

wn

48 (

20-9

7)15

80 (

22-2

00

)41

Taka

la e

t al

[71

]44

-59

(17-

86)

APA

CH

E II

45 (

10-1

00

)U

nkno

wn

154

(61-

394)

20

Gep

per

t et

al [

72]

59 (

35-8

5)SI

RS

42.8

±19

.47

96.2

±47

.327

De

Pabl

o e

t al

[73

]61

.2 (

3.2)

APA

CH

E II

4836

9852

Kay

al e

t al

[74

]57

.2 (

3.9)

AC

CP/

SCC

M40

.5 ±

4.5

923

1 ±41

.825

Shap

iro

et

al [6

]57

(19

)A

PAC

HE

III49

207

9513

And

rys

et a

l [60

]46

54.3

(8,

3-11

6.9)

68

Gia

nnak

i et

al [4

7]A

dul

t48

±13

40

AC

CP/

SCC

M =

Am

eric

an c

olle

ge o

f ch

est

phys

icia

ns/s

oci

ety

of

crit

ical

car

e m

edic

ine;

APA

CH

E =

acut

e ph

ysio

logy

and

chr

oni

c he

alth

eva

luat

ion;

PRI

SM =

ped

iatr

ic r

isk

of

mo

rtal

ity;

SIR

S =

syst

emic

infla

mm

ato

ry r

esp

ons

e sy

ndro

me.

Age

s ex

pres

sed

in y

ears

unl

ess

oth

erw

ise

stat

ed.

1 Dat

a pr

esen

ted

as

mea

n (r

ange

) o

r m

ean

± st

and

ard

dev

iati

on.

2 C

rite

ria

fro

m (

75).

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

84

5

Tab

le 3

. Lev

els

of s

olu

ble

L-se

lect

in in

neo

nate

s, c

hild

ren

and

adul

ts

sL-s

elec

tin

Stud

y [r

efer

ence

]M

ean

ag

eSe

psi

s cr

iter

ia

Hea

lth

y

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

hea

lth

y)

Sep

sis

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

sep

sis)

Neo

nate

sFi

guer

as e

t al

[49]

0-1

4 d

ays

SNA

P-II

580

(52

3-71

7)12

681 (

541-

757)

15

Kour

tis

et a

l [50

]0

-2 d

ays

Clin

ical

1155

7513

31 (

1123

-142

7)14

Gia

nnak

i et

al [4

7]A

t te

rm67

4 ±2

2340

Rebu

ck e

t al

[43]

At

term

463

(338

-557

)22

Koen

ig e

t al

[42]

0-7

day

s32

4 ±2

410

Chi

ldre

nKo

urti

s et

al [

59]

Chi

ldre

n33

56 (

2818

-389

4)10

0

Bria

sso

ulis

et

al [1

9]6.

5 (2

.8)

PRIS

M37

50 ±

321

1062

63 ±

3813

10

Ad

ults

Wei

gand

et

al [6

6]58

.7 (4

.4)

AC

CP/

SCC

M46

07

400

14

Kour

tis

et a

l [50

]A

dul

t95

0 (

700

-122

0)

75

Schl

eiff

enba

um e

t al

[23]

Ad

ult

160

0 ±

800

63

Gia

nnak

i et

al [4

7]A

dul

t93

8 ±1

8140

Rebu

ck e

t al

[43]

Ad

ult

717

(410

-822

)22

Koen

ig e

t al

[42]

Ad

ult

537

±28

9

AC

CP/

SCC

M =

Am

eric

an c

olle

ge o

f che

st p

hysi

cian

s/so

ciet

y o

f cri

tica

l car

e m

edic

ine;

PRI

SM =

ped

iatr

ic r

isk

of m

ort

alit

y; S

NA

P =

sco

re fo

r ne

ona

tal a

cute

phy

sio

logy

. Age

s ex

pres

sed

in y

ears

unle

ss o

ther

wis

e st

ated

.1 D

ata

pres

ente

d a

s m

ean

(ran

ge)

or

mea

n ±

stan

dar

d d

evia

tio

n.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

85

5

Tab

le 4

. Lev

els

of s

olu

ble

P-se

lect

in in

neo

nate

s, c

hild

ren

and

adul

ts

sP-s

elec

tin

Stud

y [r

efer

ence

]M

ean

ag

eSe

psi

s cr

iter

ia

Hea

lth

y

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

hea

lth

y)

Sep

sis

(ng

/ml)

1

Num

ber

of p

atie

nts

(sep

sis)

Neo

nate

sFi

guer

as e

t al

[49]

0-1

4 d

ays

SNA

P-II

272

(152

-288

)12

244

(170

-324

)15

Sita

ru e

t al

[48]

0 d

ays

Clin

ical

104

±71

1022

2 ±1

289

Chi

ldre

nPa

ize

et a

l [61

]2-

16PR

ISM

50 (4

4-60

)40

61 (4

7-11

9)20

Ad

ults

Mo

sad

et

al [3

2]3

SIR

S/SO

FA28

.6 ±

663

±9

176

Fijn

heer

et

al [2

9]A

dul

tSI

RS

122

±38

1039

8 ±2

03

26

Wei

gand

et

al [6

6]58

.7 (4

.4)

AC

CP/

SCC

M32

.1 ±5

.17

296

±56

14

Osm

ano

vic

et a

l [69

]A

dul

tU

nkno

wn

181 ±

4418

305

±158

9

Gep

per

t et

al [

72]

59 (

35-8

5)SI

RS

116.

9 ±3

3.4

729

1 ±22

7.427

Leo

ne e

t al

[76

]45

-47

(16-

21)

SIR

S62

±20

2612

9 ±9

811

AC

CP/

SCC

M =

Am

eric

an c

olle

ge o

f ch

est

phys

icia

ns/s

oci

ety

of

crit

ical

car

e m

edic

ine;

PRI

SM =

ped

iatr

ic r

isk

of

mo

rtal

ity;

SIR

S =

syst

emic

infla

mm

ato

ry r

esp

ons

e sy

ndro

me;

SN

AP

= sc

ore

fo

r

neo

nata

l acu

te p

hysi

olo

gy; S

OFA

= s

eque

ntia

l org

an fa

ilure

ass

essm

ent.

Age

s ex

pres

sed

in y

ears

unl

ess

oth

erw

ise

stat

ed.

1 Dat

a pr

esen

ted

as

mea

n (r

ange

) o

r m

ean

± st

and

ard

dev

iati

on.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

86

5

Tab

le 5

. Lev

els

of s

olu

ble

inte

rcel

lula

r adh

esio

n m

ole

cule

-1 in

neo

nate

s, c

hild

ren

and

adul

ts

sIC

AM

-1St

udy

[ref

eren

ce]

Mea

n a

ge

Sep

sis

crit

eria

Hea

lth

y

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

hea

lth

y)

Sep

sis

(ng

/ml)

1

Num

ber

of p

atie

nts

(sep

sis)

Neo

nate

sFi

guer

as e

t al

[49]

0-1

4 d

ays

SNA

P-II

215

(156

-274

)12

426

(394

-458

)15

Ap

ost

olo

u et

al [

58]

At

term

Clin

ical

358.

4 ±2

8.9

1071

0.7

±56

.610

Do

llner

et

al [5

2]0

-7 d

ays

Clin

ical

244

(95.

2-50

0)

2441

3 (2

55.6

-50

0)

18

Aus

tgul

en e

t al

[45]

Pre/

at te

rmC

linic

al25

8.8

(94-

500

)16

839

4.2

(197

.5-5

00

)24

Bern

er e

t al

[51]

0-3

day

sC

linic

al42

1 (29

1-45

9)35

446

(171

-534

)13

6

Edga

r et

al [

57]

0-7

day

sC

linic

al16

5 (1

30-2

90)

2734

1 (23

6-55

4)19

2

Edga

r et

al [

56]

0-7

day

sC

linic

al20

546

406

46

Chi

ldre

nA

ndry

s et

al [

60]

6-10

346.

8 (2

06.

8-48

6.8)

68

11-1

526

9 (1

84.1-

354)

90

Paiz

e et

al [

61]

2-16

PRIS

M26

0 (

240

-30

0)

4070

5 (4

00

-850

)20

Wha

len

et a

l [63

]1d

to 17

yD

oug

hty

et a

l20

5 ±2

914

595

77

Nas

h et

al [

60]

931

0 (

280

-410

)81

1530

0 (

270

-390

)

Bria

sso

ulis

et

al [1

9]6.

5 (2

.8)

PRIS

M19

9 ±9

810

172

±93

10

Ad

ults

Wei

gand

et

al [6

6]58

.7 (4

.4)

AC

CP/

SCC

M19

07

110

014

Sod

erq

uist

et

al [

70]

71 (

10-9

1)U

nkno

wn

202

(62-

392)

1545

1 (21

6-10

30)

41

Sche

rper

eel e

t al

[77

]58

.9 (

14.9

)SA

PS II

2130

63

Ho

fer

et a

l [78

]65

.9 (

12.4

)A

PAC

HE

II21

9.6

(195

.2-2

85.1)

108

444.

7 (3

30-6

65.5

)18

De

Pabl

o e

t al

[73

]61

.2 (

3.2)

APA

CH

E II

480

3611

00

52

Kay

al e

t al

[74

]57

.2 (

3.9)

AC

CP/

SCC

M20

8 ±2

0.5

986

8 ±1

3125

Shap

iro

et

al [6

]57

(19

)A

PAC

HE

III18

520

724

013

And

rys

et a

l [60

]46

140

(60

.2-2

18.4

)68

Leo

ne e

t al

[76

]45

-47

(16-

21)

SIR

S15

126

824

11

SCC

M =

Am

eric

an c

olle

ge o

f che

st p

hysi

cian

s/ s

oci

ety

of c

riti

cal c

are

med

icin

e; A

PAC

HE

= ac

ute

phys

iolo

gy a

nd c

hro

nic

heal

th e

valu

atio

n; P

RISM

= p

edia

tric

ris

k o

f mo

rtal

ity;

SA

PS =

sim

plifi

ed

acut

e ph

ysio

logy

sco

re; S

IRS

= sy

stem

ic in

flam

mat

ory

res

po

nse

synd

rom

e; S

NA

P =

sco

re fo

r ne

ona

tal a

cute

phy

sio

logy

.1 D

ata

pres

ente

d a

s m

ean

(ran

ge)

or

mea

n ±

stan

dar

d d

evia

tio

n.

2 C

rite

ria

fro

m [

75].

Age

s ex

pres

sed

in y

ears

unl

ess

oth

erw

ise

stat

ed

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

87

5

Tab

le 6

. Lev

els

of s

olu

ble

vasc

ular

cel

l adh

esio

n m

ole

cule

-1 in

neo

nate

s, c

hild

ren

and

adul

ts

sVC

AM

-1St

udy

[ref

eren

ce]

Mea

n a

ge

Sep

sis

crit

eria

Hea

lth

y

(ng

/ml)

1

Num

ber

of

pat

ien

ts (

hea

lth

y)

Sep

sis

(ng

/ml)

1

Num

ber

of p

atie

nts

(sep

sis)

Neo

nate

sFi

guer

as e

t al

[49]

0-1

4 d

ays

SNA

P-II

928

(856

-10

05)

1211

12 (

1072

-115

3)15

Aus

tgul

en e

t al

[45]

0-7

day

sC

linic

al19

40 (

110

0-3

500

)16

819

50 (

1190

-349

5)24

Chi

ldre

nA

ndry

s et

al [

60]

6 -1

5 ye

ars

590

.8 (

359.

6-82

2.0

)68

Paiz

e et

al [

61]

2.16

PRIS

M60

0 (

390

-790

)40

1550

(13

60-1

910

)20

Kru

eger

et

al [6

2]3.

5 (0

.2-1

6)A

CC

P/SC

CM

766

(644

-915

)22

1239

(92

8-16

15)

22

Wha

len

et a

l [63

]1d

to 17

yD

oug

hty

et a

l23

1 ±46

1460

077

Nas

h et

al [

64]

9 ye

ars

790

(58

0-1

060

)81

15 y

ears

780

(420

-10

00

)

Ad

ults

Pres

terl

et

al [6

5]51

APA

CH

E II

545

(374

-829

)20

2633

20

Kna

pp e

t al

[68]

51-5

5 (2

1-96

)A

PAC

HE

III56

9 ±9

815

1395

±80

128

Sod

erq

uist

et

al [

70]

71 (

10-9

1)U

nkno

wn

533

(354

-10

18)

1511

73 (

525-

350

0)

41

Ho

fer

et a

l [78

]65

.9 (

12.4

)A

PAC

HE

II15

24.7

(99

1.2-

2038

)10

811

47.9

(88

3.5-

2074

.4)

18

De

Pabl

o e

t al

[73

]61

.2 (

3.2)

APA

CH

E II

890

4816

00

52

Shap

iro

et

al [6

]57

(19

)A

PAC

HE

III10

5049

1550

95

And

rys

et a

l [60

]46

743

(338

-114

8)68

Leo

ne e

t al

[76

]45

-47

(16-

21)

SIR

S45

8 ±1

2326

160

4 ±9

4011

AC

CP/

SCC

M =

Am

eric

an c

olle

ge o

f ch

est

phys

icia

ns/

soci

ety

of

crit

ical

car

e m

edic

ine;

APA

CH

E, a

cute

phy

sio

logy

and

chr

oni

c he

alth

eva

luat

ion;

PRI

SM =

ped

iatr

ic r

isk

of

mo

rtal

ity;

SIR

S =

syst

emic

infla

mm

ato

ry r

esp

ons

e sy

ndro

me;

SN

AP

= sc

ore

for

neo

nata

l acu

te p

hysi

olo

gy.

1 Dat

a pr

esen

ted

as

mea

n (r

ange

) o

r m

ean

± st

and

ard

dev

iati

on.

2 C

rite

ria

fro

m [

75].

Age

s ex

pres

sed

in y

ears

unl

ess

oth

erw

ise

stat

ed.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

88

5

Similarly, in a large pediatric ICU study on microcirculatory dysfunction in meningococcal sepsis

in children, levels of sE-selectin, sVCAM-1 and sICAM-1, but not sP-selectin, were significantly

increased in septic patients but negatively correlated with the degree of microcirculatory

dysfunction (a measure of sepsis severity), as assessed by sublingual imaging [61].

Adults

Generally, basal levels of soluble adhesion molecules in adults are similar to or somewhat lower

than those of neonates and children (Tables 2, 3, 4, 5 and 6 and Figure 3). All molecules show

increase during sepsis, with sICAM-1 and sP-selectin exhibiting the greatest increases compared

with neonates and children. Age group stratification in levels of soluble adhesion molecules in

adults is limited. Rudloff and colleagues determined sICAM-1 levels in healthy adults between 18

and 65 years old and reported no age-dependent differences [79].

As discussed above, in a large number of clinical sepsis studies in adults, higher levels of

soluble adhesion molecules were generally related to severity of disease and mortality, although

statistically significant correlations could not be made [5,6,15-17,37,65-74,76-82]. Some studies imply

an alternate interpretation of these levels. For example, clinical studies have demonstrated that

septic adults with modest levels of soluble adhesion molecules (putatively reflecting inadequate/

aberrant shedding) had poorer outcome and higher mortality than those with the highest levels

[22,83]. Donnelly and colleagues found that critically ill patients with lower levels of sL-selectin had

a higher chance of developing adult respiratory distress syndrome [83], and Seidelin and colleagues

found that worse outcomes in septic patients correlated with lesser increases in sL-selectin levels

[22]. Interestingly, one experimental study found significantly decreased leukocyte–endothelial

interaction in a murine cecal ligation model of sepsis upon addition of exogenous sL-selectin into

the circulation at levels comparable with those found in septic adults [27].

ADHESION MOLECULE SHEDDASES IN SEPSIS: A DELICATE BALANCE The levels of adhesion molecules are an indirect result of protein cellular expression levels and

a direct result of the proteolytic activity of sheddases. Thus, as discussed above (for example, see

[42-44], expression and shedding activities can both independently contribute to overall levels

of soluble adhesion molecules in circulation. Several studies have independently assessed levels

of circulating sheddases and sheddase antagonists (that is, tissue inhibitor metalloproteinases) in

clinical and experimental sepsis in efforts to clarify their contribution to pathology. However, so

far these have yielded diverse and inconsistent results showing varied correlation of levels with

protection and pathology [84-88], suggesting a delicate balance is required.

The sensitive relationship between levels/activity of sheddases and outcome/effects in

the host during sepsis is best reflected by studies from Long and colleagues investigating

the role of ADAM-17 in L-selectin shedding in murine sepsis [12,89]. They found that ADAM-17

in mice acts as a homeostatic (rheostat) molecule to control their neutrophil infiltration at sites

of inflammation by regulating surface density of L-selectin. Low ADAM-17 activity results in little

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

89

5

L-selectin shedding and too much neutrophil infiltration with subsequent collateral tissue damage.

However, excessive activity of ADAM-17 promotes excessive L-selectin shedding and subsequently

impairs neutrophil infiltration, which is needed to clear inflammation and infection.

Finally, evidence of age-related changes in sheddases and sheddase antagonists (that is,

tissue inhibitor metalloproteinases) has been observed. It is interesting to speculate that these

differences could partially underlie the observed age-related discrepancies in levels of soluble

adhesion molecules [90,91].

CONCLUSIONIncreased levels of soluble adhesion molecules generally correlate well with the presence of sepsis

in neonates, children and adults. However, their levels are still poorly predictive of sepsis severity

scores, outcome and mortality. Our review raises important issues that need further attention,

including age-related discrepancies in soluble adhesion molecule levels and even basic questions

about whether these should correlate positively or negatively with mortality.

First, there is a well-articulated hypothesis (and significant experimental support) that

shedding is indeed principally a homeostatic process that works to reduce inflammation and

promote resolution of inflammation (Figure 4). This is thought to act at two complementary

levels: removal of adhesion molecules from cell surfaces directly reduces the ability for cell–cell

interaction; and the resulting soluble isoforms serve as competitive antagonists (or decoy ligands)

for the remaining cell surface adhesion molecules.

Second, there is substantial evidence that disruption of shedding during sepsis, resulting

in substantially lower levels of soluble adhesion molecules (that is, retention of elevated cell

surface adhesion molecule levels), could lead to exacerbation of inflammation or promotion of

mortality (Figure 4C). Thus, as illustrated in Figure 4, there are many points during the progression

of an inflammatory response whereby the functional significance (with respect to the severity

of the underlying inflammation) of a given level of soluble adhesion molecule varies greatly. In

addition, levels of sheddases are also altered in human and experimental sepsis, suggesting putative

functional contribution for these changes in regulating disease progression. The interaction of

adhesion molecules and sheddases in the dynamic microenvironment of the cellular surfaces

implicates a strong interdependence of these molecules. However, to our knowledge, no studies

exist that combine the assessment of both adhesion molecules and their sheddases to assess

clinical outcome. Thus, a novel and potentially critical opportunity to enhance clinical efficacy of

soluble adhesion molecules exists that remains untapped. Such a combinatorial approach might be

particularly useful for improving both sepsis diagnosis (which is greatly needed for the challenging

situations in the febrile neonate or extreme older person) and our ability to track efficacy of

therapies. Furthermore, serial assessment of the combination of these markers might even be

effective in determining morbidity or mortality risk. Finally, it is interesting to consider whether

regulation of sheddases and adhesion molecules might be regulated in sepsis at the epigenetic

level, potentially offering additional ways to assess disease severity and predict outcomes [92].

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

90

5

Additionally, the age-related and comorbidity-related heterogeneity in levels of soluble

adhesion molecules, as well as sheddases, in healthy individuals and septic patients could be an

expression of a different basal state, as well as different responsiveness in sepsis, potentially leading

to discrepancies in pathophysiology and disease progression between neonates, children or

adults. Interestingly, epidemiological research shows a biphasic pattern in age-related difference

in incidence and mortality [2-4]. The incidene of neonatal sepsis is 1 to 8 per 1,000 live births with

mortality rates of 16%. These rates decrease during childhood (0.2/1,000 children, mortality 10%)

and then increase in adults (26.2/1,000 in those over 85 years old, mortality 38.4%) [2-4]. A direct

correlation between these rates and levels of soluble adhesion molecules remains speculative, but

further attention to this could provide new insights.

In conclusion, while predictive properties of soluble adhesion molecules have been researched

intensively, their levels are still poorly predictive of sepsis outcome and mortality. We propose

two novel directions for improving clinical utility of soluble adhesion molecules: the combined

simultaneous analysis of levels of adhesion molecules and their sheddases; and taking age-

related discrepancies into account. Additional investigation of these issues may provide better

understanding of the pathophysiology of sepsis and increased usefulness of soluble adhesion

molecules as diagnostic and predictive biomarkers.

Abbreviations

ADAM = a disintegrin and metalloproteinase

ICAM-1 = intercellular adhesion molecule-1

sE-selectin = soluble E-selectin

sICAM-1 = soluble intercellular adhesion molecule-1

sL-selectin = soluble L-selectin

sP-selectin = soluble P-selectin

sVCAM-1 = soluble vascular cell adhesion molecule-1

VCAM-1 = vascular cell adhesion molecule-1

Competing Interests

The authors declare that they have no competing interests.

Author Contributions

RZ performed the literature search. RZ, RM, NIS, TWK, FBP and CVC helped draft and revise

the manuscript. RZ and CVC prepared the figures. All authors helped to draft the manuscript. All

authors read and approved the final manuscript.

Acknowledgments

R.Z. was supported by stipends from the Tergooi Hospitals, Blaricum, the Drie Lichten foundation,

and the Ter Meulen Fund, Royal Netherlands Academy of Arts and Sciences and the IPRF Early

Investigators Exchange Program Award of the European Society for Pediatric Research. C.V.C. was

supported by a grant from the NIH (HL104006).

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

91

5

Idealized Homeostatic In ammatory Response

Abberant Shedding & Sustained In ammation

Cell Surface Adhesion Molecules Soluble/Shed Adhesion Molecules

seluceloM noisehdA f o sl eveL

Overall Level of In ammation

seluceloM noisehdA f o sl eveL

sel ucel oM noi sehdA f o sl eveL

In ammation Onset In ammation Resolution

Exaggerated In ammatory Response

A

B

C

Time

Figure 4. Changes in cell-surface and circulating soluble adhesion molecules during progression of

inflammatory responses. A: Idealized Homeostatic Inflammatory Response. Schematic shows hypothetical

increase in cell surface adhesion molecules (blue line) associated with onset of inflammation and subsequent

decreases in them during resolution of inflammation. Shedding and accumulation of circulating soluble

adhesion molecules similarly rise and fall (green line), but with a lag in time. Points of highest levels of

cell-surface adhesion molecules (and not necessarily soluble adhesion molecules) should correlate with

the greatest levels of overall inflammation and propensity for collateral tissue damage (red shading);

B: Exaggerated Inflammatory and Response. Schematic as in A illustrates a more severe inflammation and

greater initial cell surface adhesion molecule expression and subsequently greater levels of soluble adhesion

molecule production; C: Aberrant Shedding and Sustained Inflammation. Schematic as in A illustrates

how a hypothetical deficiency in shedding during inflammation could lead to low levels of soluble

adhesion molecules, leaving cell-surface adhesion molecules elevated, and allowing for heightened and

sustained inflammation.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

92

5

REFERENCES1. Dellinger RP, Carlet JM, Masur H, Gerlach H,

Calandra T, Cohen J, Gea-Banacloche J, Keh D,

Marshall JC, Parker MM, Ramsay G, Zimmerman

JL, Vincent JL, Levy MM; Surviving Sepsis

Campaign Management Guidelines Committee.

Surviving Sepsis Campaign guidelines for

management of severe sepsis and septic shock.

Crit Care Med 2004, 32(3):858-73.

2. Lukacs SL, Schoendorf KC, Schuchat A.

Trends in sepsis –related neonatal mortality

in the United States. 1985-1998. Pediatr Infect

Dis J 2004, 23(7):599-603.

3. Stoll BJ, Hansen NI, Sánchez PJ, Faix RG,

Poindexter BB, Van Meurs KP, Bizzarro MJ,

Goldberg RN, Frantz ID 3rd, Hale EC, Shankaran

S, Kennedy K, Carlo WA, Watterberg KL, Bell EF,

Walsh MC, Schibler K, Laptook AR, Shane AL,

Schrag SJ, Das A, Higgins RD; Eunice Kennedy

Shriver National Institute of Child Health and

Human Development Neonatal Research

Network. Early onset neonatal sepsis: the burden

of group B-streptococcal and E.coli disease

continues. Pediatrics 2011, 127(5):817-826.

4. Angus DC, Linde-Zwirble WT, Lidicker

J, Clermont G, Carcillo J, Pinsky MR.

Epidemiology of severe sepsis in the United

States: analysis of incidence, outcome,

and associated costs of care. Crit Care

Med 2001, 29(7):1303-1310.

5. Aird WC. The role of the endothelium in

severe sepsis and multiple organ dysfunction

syndrome. Blood 2003, 101(10):3765-3777.

6. Shapiro NI, Schuetz P, Yano K, Sorasaki M,

Parikh SM, Jones AE, Trzeciak S, Ngo L, Aird WC.

The association of endothelial cell signalling,

severity of illness, and organ dysfunction in

sepsis. Crit Care 2010, 14(5):R182.

7. Carman CV, Springer TA. Trans-cellular

migration: cell-cell contacts get intimate. Curr

Opin Cell Biol 2008, 20(5):533-4.

8. Springer TA. Adhesion receptors of

the immune system. Nature 1990, 346:425-433.

9. Springer TA. Traffic signals for lymphocyte

recirculation and leukocyte emigration:

the multi-step paradigm. Cell 1994, 76:301-314.

10. Garton KJ, Gough PJ, Raines EW. Emerging roles

for ectodomain shedding in the regulation

of inflammatory responses. J Leukoc

Biol 2006, 79(6):1105-16.

11. Xing K, Murthy S, Liles WC, Singh JM. Clinical

utility of biomarkers of endothelial activation in

sepsis-a systematic review. Crit Care 2012, 16:R7.

12. Long C, Wang Y, Herrera AH, Horiuchi K,

Walcheck B. In vivo role of leukocyte ADAM17

in the inflammatory and host responses

during E. coli-mediated peritonitis. J Leukoc

Biol 2010, 87(6):1097-101.

13. Ley K, Laudanna C, Cybulsky MI, Nourshargh

S. Getting to the site of inflammation:

the leukocyte adhesion cascade updated. Nat

Rev Immunol 2007, 7(9):678-89.

14. Harrington EO, Stefanec T, Newton J,

Rounds S. Release of soluble E-selectin from

activated endothelial cells upon apoptosis.

Lung 2006, 184(5):259-66.

15. Pigot R, Dillon LP, Hemingway IH, Gearing

AJ. Soluble forms of E-selectin, ICAM-1 and

VCAM-1 are present in the supernatants of

cytokine activated cultured endothelial cells.

Biophys Res Commun 1992, 187(2):584–589.

16. Newman W, Beall LD, Carson CW, Hunder GG,

Graben N, Randhawa ZI, Gopal TV, Wiener-

Kronish J, Matthay MA. Soluble E-selectin is

found in supernatants of activated endothelial

cells and is elevated in the serum of patients with

septic shock. J Immunol 1993, 150(2):644 -654.

17. Cummings CJ, Sessler CN, Beall LD, Fisher BJ,

Best AM, Fowler AA 3rd. Soluble E-selectin

levels in sepsis and critical illness. Correlation

with infection and hemodynamic dysfunction.

Am J Respir Crit Care Med 1997, 156(2):431-437.

18. Smith CW. Potential significance of circulating

E-selectin. Circulation 1997, 15;95(8).

19. Briassoulis G, Papassotiriou I, Mavrikiou M,

Lazaropoulou C, Margeli A. Longitudinal

course and clinical significance of TGF-beta1,

sL- and sE-Selectins and sICAM-1 levels

during severe acute stress in children. Clin

Biochem. 2007;40(5-6):299-304.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

93

5

20. Smalley DM, Ley K. L-selectin: mechanisms

and physiological significance of ectodomain

cleavage. J Cell Mol Med 2005, 9(2):255-66.

21. Venturi GM, Tu L, Kadono T, Khan AI,

Fujimoto Y, Oshel P, Bock CB, Miller AS,

Albrecht RM, Kubes P, Steeber DA, Tedder

TF. Leukocyte migration is regulated

by L-selectin endoproteolytic release.

Immunity 2003,19(5):713-24.

22. Seidelin JB, Nielsen OH, Strøm J. Soluble

L-selectin levels predict survival in sepsis.

Intensive Care Med 2002, 28(11):1613-8.

23. Schleiffenbaum B, Spertini O, Tedder TF.

Soluble L-selectin is present in human plasma

at high levels and retains functional activity. J

Cell Biol 1992, 119(1):229-238.

24. Ferri LE, Pascual J, Seely AJ, Chaudhury P,

Christou NV. Soluble L-selectin attenuates

tumor necrosis factor-β-mediated leukocyte

adherence and vascular permeability:

A protective role for elevated soluble L-selectin

in sepsis. Crit Care Med 2002, 30(8):1842-1847.

25. Ferri LE, Swartz D, Christou NV. Soluble

L-selectin at levels present in septic patients

diminishes leukocyte-endothelial cell

interactions in mice in vivo: a mechanism for

decreased leukocyte delivery to remote sites

in sepsis. Crit Care Med 2001, 29(1):117-22.

26. Ferri LE, Chia S, Benay C, Giannias B, Christou

NV. L-selectin shedding in sepsis limits

leukocyte mediated microvascular injury at

remote sites. Surgery 2009, 145(4):384-391.

27. Easton AS, Dorovini-Zis K. The kinetics,

function, and regulation of P-selectin

expressed by human brain microvessel

endothelial cells in primary culture. Microvasc

Res 2001, 62(3):335-45.

28. Zarbock A, Polanowska-Grabowska RK, Ley

K. Platelet-neutrophil interactions: linking

haemostasis and inflammation. Blood

Rev 2007, 21(2):99-111.

29. Fijnheer R, Frijns CJ, Korteweg J, Rommes

H, Peters JH, Sixma JJ, Nieuwenhuis HK.

The origin of P-selectin as a circulating plasma

protein. Thromb Haemost 1997, 77(6):1081-5.

30. Chen AY, Ha JN, Delano FA, Schmid-Schönbein

GW. Receptor cleavage and P-selectin-

dependent reduction of leukocyte adhesion in

the spontaneously hypertensive rat. J Leukoc

Biol 2012, 92(1):183-94.

31. Füth R, Dinh W, Nickl W, Bansemir L, Barroso

MC, Bufe A, Sause A, Scheffold T, Krahn T,

Ellinghaus P, Lankisch M. Soluble P-selectin

and matrix metalloproteinase 2 levels are

elevated in patients with diastolic dysfunction

independent of glucose metabolism

disorder or coronary artery disease. Exp Clin

Cardiol 2009, 14(3):76-79.

32. Mosad E, Elsayh KI, Eltayeb AA. Tissue factor

pathway inhibitor and P-selectin as markers

of sepsis-induced non-overt disseminated

intravascular coagulopathy. Clin Appl Thromb

Hemost 2011, 17(1):80-87.

33. Tsakadze NL, Sithu SD, Sen U, English WR,

Murphy G, D’Souza SE. Tumor necrosis factor-

alpha-converting enzyme (TACE/ADAM-17)

mediates the ectodomain cleavage of

intercellular adhesion molecule-1 (ICAM-1). J

Biol Chem 2006, 281(6): 3157-3164.

34. Sultan S, Gosling M, Nagase H, Powell

JT. Shear stress-induced shedding of

soluble intercellular adhesion molecule-1

from saphenous vein endothelium. FEBS

Lett 2004, 564(1-2):161-5.

35. Fiore E, Fusco C, Romero P, Stamenkovic

I. Matrix metalloproteinase 9 (MMP-9/

gelatinase B) proteolytically cleaves ICAM-1

and participates in tumor cell resistance

to natural killer cell-mediated cytotoxicity.

Oncogene 2002, 21(34):5213-23.

36. Champagne B, Tremblay P, Cantin A, St Pierre Y.

Proteolytic cleavage of ICAM-1 by human neutrophil

elastase. J Immunol 1998, 161(11):6398-405.

37. Griensven van M, Probst C, Müller K, Hoevel P,

Pape HC. Leukocyte-endothelial interactions

via ICAM-1 are detrimental in polymicrobial

sepsis. Shock 2006, 25(3):254-259.

38. Shapiro NI, Yano K, Sorasaki M, Fischer C, Shih

SC, Aird WC. Skin biopsies demonstrate site-

specific endothelial activation in mouse models

of sepsis. J Vasc Res 2009, 46(5):495-502.

39. Lemaire LC, de Kruif MD, Giebelen IA, van

Zoelen MA, van’t Veer C, van der Poll T.

Differential dose-dependent effects of

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

94

5

prednisolone on shedding of endothelial

adhesion molecules during human

endotoxemia. Immunol Lett 2008, 21(2):93-6.

40. Singh RJ, Mason JC, Lidington EA, Edwards DR,

Nuttall RK, Khokha R, Knauper V, Murphy G,

Gavrilovic J. Cytokine stimulated vascular cell

adhesion molecule-1 (VCAM-1) ectodomain

release is regulated by TIMP-3. Cardiovasc

Res 2005, 67(1):39-49.

41. Newby AC. Studying mechanisms underlying

shedding of endothelial membrane proteins

could help patients at risk for myocardial

infarction. Cardiovasc Res 2005, 67(1):4-5.

42. Koenig JM, Simon J, Anderson DC, Smith

E, Smith CW. Diminished soluble and total

cellular L-selectin in cord blood is associated

with its impaired shedding from activated

neutrophils. Ped Res 1996, 39(4):616-621.

43. Rebuck N, Gibson A, Finn A. Neutrophil adhesion

molecules in term and premature infants:

normal or enhanced leucocyte integrins but

defective L-selectin expression and shedding.

Clin Exp Immunol 1995, 101(1):183-189.

44. Török C, Lundahl J, Hed J, Lagercrantz H.

Diversity in regulation of adhesion molecules

(Mac-1 and L-selectin) in monocytes and

neutrophils from neonates and adults. Arch

Dis Child 1993, 68:561-565.

45. Austgulen R, Arntzen KJ, Haereid PE, Aag S,

Døllner H. Infections in neonates delivered

at term are associated with increased

serum levels of ICAM-1 and E-selectin. Acta

Pediatr 1997, 86(3):274-280 .

46. Levy O. Innate immunity of the newborn: basic

mechanisms and clinical correlates. Nat Rev

Immunol 2007, 7(5):379-90.

47. Giannaki G, Rizos D, Xyni K, Sarandakou A,

Protonotariou E, Phocas I, Creatsas G. Serum

soluble E-and L-selectin in the very early neonatal

period. Early Hum Develop 2000, 60(2):149-155

48. Sitaru AG, Speer CP, Holzhauer S, Obergfell

A, Walter U, Grossmann R. Chorioamnionitis

is associated with increased CD40L

expression on cord blood platelets. Thromb

Haemost 2005, 94(6):1219-1223

49. Figueras-Aloy J, Gómez-López L, Rodríguez-

Miguélez JM, Salvia-Roiges MD, Jordán-

García I, Ferrer-Codina I, Carbonell-Estrany X,

Jiménez-González R. Serum Soluble ICAM-1,

VCAM-1, L-selectin and P-selectin levels

as markers of infection and their relation

to clinical severity in neonatal sepsis. Am J

Perinat 2007, 24(6):331-338.

50. Kourtis AP, Lee FK, Stoll BJ. Soluble L-selectin,

a marker of immune activation, in neonatal

infection. Clin Immunol 2003, 109(2):224-228.

51. Berner R, Niemeyer CM, Leititis JU, Funke A,

Schwab C, Rau U, Richter K, Tawfeek MS, Clad

A, Brandis M. Plasma levels and gene expression

of granulocyte colony-stimulating factor,

tumor necrosis factor –(alpha), interleukin

(IL)-1(beta), IL-6, IL-8, and soluble intercellular

adhesion molecule-1 in neonatal early onset

sepsis. Ped Res 1998, 44(4):469-477.

52. Dollner H, Vatten L, Austgulen R. Early

diagnostic markers for neonatal sepsis:

comparing c-reactive protein, interleukin-6,

soluble tumor necrosis factor receptors

and soluble adhesion molecules. J Clin

Epid 2001, 54(12):1251-1257.

53. Hansen AB, Verder H, Staun-Olsen P. Soluble

intercellular adhesion molecule and c-reactive

protein as early markers of infection in

newborns. J Perinat Med 2000, 28(2):97-103.

54. Edgar JDM. Serum sICAM-1 levels in

the diagnosis of neonatal infection. Ped

Res 1998, 44:430.

55. Edgar JDM, Wilson DC, McMillan SA, Crockard

AD, Halliday MI, Gardiner KR, Rowlands BJ,

Halliday HL, McNeill TA. Predictive value of

soluble immunological mediators in neonatal

infection. Clin Sci 1994, 87(2):165.

56. Edgar JDM, Gabriel V, Craig A, Wheeler D,

Thomas M, Grant J. A low serum sICAM-1

level may assist in the exclusion of neonatal

infection. Biol Neonate 2002, 81(2):105-108.

57. Edgar JDM, Gabriel V, Gallimore JR,

McMillan SA, Grant J. A prospective study

of the sensitivity, specificity and diagnostic

performance of soluble intercellular adhesion

molecule 1, highly sensitive C-reactive

protein, soluble E-selectin and serum amyloid

A in the diagnosis of neonatal infection. BMC

Pediatr 2010, 16:10-22.

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

95

5

58. Apostolou M, Dimitriou H, Kaleyias J,

Perdikogianni C, Stiakaki E, Costalos C, Kalmanti

MC. Levels of soluble ICAM-1 in premature and

full-term neonates with infection. Mediators

Inflamm 2002, 11(2):95–98.

59. Kourtis AP, Nesheim SR, Thea D, Ibegbu C,

Nahmias AJ, Lee FK. Correlation of virus

load and soluble L-selectin, a marker of

immune activation, in pediatric HIV infection.

AIDS 2000, 14(16): 2429-2436.

60. Andrys C, Pozler O, Krejsek J, Derner V,

Drahosová M, Kopecký O. Serum soluble

adhesion molecules (sICAM-1, sVCAM and

E-selectin) in healthy school aged children and

adults. Acta Medica 2000, 43(3):103-106.

61. Paize F, Sarginson R, Makwana N, Baines PB,

Thomson AP, Sinha I, Hart CA, Riordan A,

Hawkins KC, Carrol ED, Parry CM. Changes

in the sublingual microcirculation and

endothelial adhesion molecules during

the course of severe meningococcal disease

treated in the paediatric intensive care unit.

Intensive Care Med 2012, 38(5):863-871.

62. Krueger M, Heinzmann A, Nauck M. Adhesion

molecules in pediatric intensive care patients

with organ dysfunction syndrome. Int Care

med 2007, 33(2):359-363 .

63. Whalen MJ, Doughty LA, Carlos TM, Wisniewski

SR, Kochanek PM, Carcillo JA. Intercellular

adhesion molecule-1 and vascular cell adhesion

molecule-1 are increased in the plasma of

children with sepsis-induced multiple organ

failure. Crit Care Med 2000, 28(7):2600-2607.

64. Nash MC, Wade AM, Shah V, Dillon MJ.

Normal levels of soluble E-selectin, soluble

intercellular adhesion molecule-1 (sICAM-1),

and soluble vascular adhesion molecule-

1 (sVCAM-1) decrease with age. Clin Exp

Immunol 1996, 103(1):167-170.

65. Presterl E, Lassnigg A, Mueller-Uri P, El-

Menyawi I, Graninger W. Cytokines in sepsis

due to Candida albicans and in bacterial sepsis.

Eur Cytokine Netw 1999, 10(3):423-30.

66. Weigand MA, Schmidt H, Pourmahmoud M,

Zhao Q, Martin E, Bardenheuer HJ. Circulating

intercellular adhesion molecule-1 as an early

predictor of hepatic failure in patients with septic

shock. Crit Care Med 1999, 27(12):2656-2661.

67. Hynninen M, Valtonen M, Markkanen H, Vaara

M, Kuusela P, Jousela I, Piilonen A, Takkunen

O. Interleukin 1 receptor antagonist and

E-selectin concentrations: a comparison in

patients with severe acute pancreatitis and

severe sepsis. J Crit Care 1999, 14(2):63-68.

68. Knapp S, Thalhammer F, Locker GJ, Laczika

K, Hollenstein U, Frass M, Winkler S, Stoiser

B, Wilfing A, Burgmann H. Prognostic value of

MIP-1 alpha, TGF-beta 2, sELAM-1, and sVCAM-1

in patients with gram-positive sepsis. Clin

Immunol Immunopathol 1998, 7(2):139-144.

69. Osmanovic N, Romijn FP, Joop K, Sturk A,

Nieuwland R. Soluble selectins in sepsis:

microparticle-associated, but only to a minor

degree. Thromb Haemost. 2000 84(4):731-732.

70. Soderquist B, Sundqvist KG, Vikerfors T.

Adhesion molecules E-selectin, intercellular

adhesion molecules-1 (ICAM-1) and vascular

cell adhesion molecule-1 (VCAM-1) in sera

from patients with Staphylococcus aureus

bacteremia with or without endocarditis. Clin

Exp Immunol 1999, 118(3):408-411.

71. Takala A, Jousela I, Jansson SE, Olkkola KT,

Takkunen O, Orpana A, Karonen SL, Repo H.

Markers of systemic inflammation predicting

organ failure in community-acquired septic

shock. Clin Sci (Lond) 1999, 97(5):529-538.

72. Geppert A, Zorn G, Karth GD, Haumer M,

Gwechenberger M, Koller-Strametz J, Heinz G,

Huber K, Siostrzonek P. Soluble selectins and

the systemic inflammatory response syndrome

after successful cardiopulmonary resuscitation.

Crit Care Med 2000, 28(7):2360-2365.

73. De Pablo R, Monserrat J, Reyes E, Diaz D,

Rodriguez-Zapata M, de la Hera A, Prieto

A, Alvarez-Mon M. Circulating sICAM-1

and sE-Selectin as biomarker of infection

and prognosis in patients with systemic

inflammatory response syndrome. Eur J Intern

Med 2013, 24(2):132-138.

74. Kayal S, Jaïs JP, Aguini N, Chaudière J,

Labrousse J. Elevated circulating E-selectin,

intercellular adhesion molecule 1, and

von Willebrand factor in patients with

SOLU

BLE AD

HESIO

N M

OLEC

ULES IN

SEPSIS

96

5

severe infection. Am J Respir Crit Care

Med 1998, 157(3): 776-784

75. Doughty LA, Kaplan SS, Carcillo JA.

Inflammatory cytokine and nitric oxide

responses in pediatric sepsis and organ failure.

Crit Care Med 1996, 24:1137-1143

76. Leone M, Boutière B, Camoin-Jau L, Albanèse

J, Horschowsky N, Mège JL, Martin C, Dignat-

George F. Systemic endothelial activation

is greater in septic than in traumatic-

hemorrhagic shock but does not correlate

with endothelial activation in skin biopsies.

Crit Care Med 2002, 30:808-814

77. Scherpereel A, Depontieu F, Grigoriu B,

Cavestri B, Tsicopoulos A, Gentina T, Jourdain

M, Pugin J, Tonnel AB, Lassalle P. Endocan,

a new endothelial marker in human sepsis. Crit

Care Med 2006, 34(2):532-537.

78. Hofer S, Brenner T, Bopp C, Steppan J,

Lichtenstern C, Weitz J, Bruckner T, Martin

E, Hoffmann U, Weigand MA. Cell death

serum biomarkers are early predictors for

survival in severe septic patients with hepatic

dysfunction. Crit Care. 2009;13(3):R93.

79. Rudloff S, Thomas C, Kunz C. Variations of

soluble intercellular cell adhesion molecule.

Eur J Med Res 1995, 1(3):171-172.

80. Gearing JH, Newman W. Circulating

adhesion molecules in disease. Immunol

Today 1993, 14(10): 506-12.

81. Pierrakos C, Vincent JL. Sepsis biomarkers:

a review. Crit Care Med 2010, 14(1):15.

82. Schuetz P, Jones AE, Aird WC, Shapiro NI.

Endothelial cell activation in emergency

department patients with sepsis-related

and non-sepsis related hypotension.

Shock 2011, 36(2):104-108.

83. Donnelly SC, Haslett C, Robertson CE, Carter

DC, Ross JA, Grant IS, Tedder TF. Role of selectins

in development of adult respiratory distress

syndrome. The Lancet 1994, 344:8917:215-219.

84. Hoffmann U, Brueckmann M, Borggrefe M.

Matrix metalloproteinases and their inhibitors:

promising novel biomarkers in severe sepsis?

Crit Care 2009, 13(6):1006.

85. Lauhio A, Hästbacka J, Pettilä V, Tervahartiala

T, Karlsson S, Varpula T, Varpula M, Ruokonen

E, Sorsa T, Kolho E. Serum MMP-8, -9 and

TIMP-1 in sepsis: high serum levels of MMP-8

and TIMP-1 are associated with fatal outcome

in a multicentre, prospective cohort study.

Hypothetical impact of tetracyclines.

Pharmacol Res 2011, 64(6):590-4.

86. Lorente L, Martín MM, Labarta L, Díaz C, Solé-

Violán J, Blanquer J, Orbe J, Rodríguez JA,

Jiménez A, Borreguero-León JM, Belmonte

F, Medina JC, Llimiñana MC, Ferrer-Agüero

JM, Ferreres J, Mora ML, Lubillo S, Sánchez

M, Barrios Y, Sierra A, Páramo JA. Matrix

metalloproteinase-9, -10, and tissue inhibitor

of matrix metalloproteinases-1 blood levels as

biomarkers of severity and mortality in sepsis.

Crit Care 2009, 13(5):R158.

87. Yazdan-Ashoori P, Liaw P, Toltl L, Webb B,

Kilmer G, Carter DE, Fraser DD. Elevated

plasma matrix metalloproteinases and their

tissue inhibitors in patients with severe sepsis.

J Crit Care 2011;26(6):556-65.

88. Belaaouaj A, McCarthy R, Baumann M, Gao

Z, Ley TJ, Abraham SN, Shapiro SD. Mice

lacking neutrophil elastase reveal impaired

host defense against gram negative bacterial

sepsis. Nat Med 1998, 4(5):615-8.

89. Long C, Hosseinkhani MR, Wang Y, Sriramarao

P, Walcheck B. ADAM17 activation in

circulating neutrophils following bacterial

challenge impairs their recruitment. J Leukoc

Biol 2012, 92(3):667-72.

90. Bonnema DD, Webb CS, Pennington WR,

Stroud RE, Leonardi AE, Clark LL, McClure CD,

Finklea L, Spinale FG, Zile MR. Effects of age on

plasma matrix metalloproteinases (MMPs) and

tissue inhibitor of metalloproteinases (TIMPs).

J Card Fail 2007, 13(7):530-540.

91. Tayebjee MH, Lip GY, Blann AD, Macfadyen

RJ. Effects of age, gender, ethnicity, diurnal

variation and exercise on circulating levels of

matrix metalloproteinases (MMP)-2 and -9,

and their inhibitors, tissue inhibitors of matrix

metalloproteinases (TIMP)-1 and -2. Thromb

Res 2005, 115(3):205-210.

92. Graves BT, Munro CL. Epigenetics in

Critical Illness: A New Frontier. Nurs Res

Pract 2013, 2013:503686.

6 Early Onset Sepsis in Surinamese Newborns is Not Associated with

Elevated Serum Levels of Endothelial Cell Adhesion Molecules and

Their Shedding Enzymes

Rens Zonneveld, Rianne M. Jongman, Amadu Juliana, Grietje Molema, Matijs van Meurs, Frans B. Plötz

Submitted

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

100

6

ABSTRACTSObjectives

Leukocyte-endothelial interactions play a pivotal part in sepsis pathophysiology. During sepsis

in adults, endothelial cell adhesion molecules (CAMs) orchestrate these interactions and their

soluble isoforms (sCAMs) are released into the vasculature by enzymes called sheddases. We

hypothesized that sCAMs and sheddases circulate at higher levels in blood culture positive early

onset sepsis (EOS) in newborns and that they are useful as biomarkers for EOS.

Materials and Methods

Soluble CAMs sP-selectin, sE-selectin, vascular cell adhesion molecule-1 (sVCAM-1), intercellular

adhesion molecule-1 (sICAM-1) and platelet and endothelial cell adhesion molecule-1

(sPECAM-1), sheddases matrix metalloproteinase-9 (MMP-9) and neutrophil elastase (NE),

and sheddase antagonist tissue-inhibitor of metalloproteinases-1 (TIMP-1) were measured

simultaneously in serum of 71 Surinamese newborns suspected of EOS and 20 healthy newborns,

all included within 72 hours after birth.

Results

Six (8.5%) newborns had a positive blood culture. At start of antibiotic treatment and after 48-72

hours no differences were found in levels of sCAMs and sheddases between blood culture positive

EOS and controls. Median sP-selectin levels associated with higher postnatal age (Spearman’s

rho -0.21; P=0.03).

Conclusions

Our data indicate that endothelial CAM shedding is not increased in EOS and that levels of sCAMs

and sheddases remain unchanged in early life in newborns. Therefore, these markers have limited

clinical utility as biomarkers for EOS.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

101

6

INTRODUCTIONEarly onset sepsis (EOS) in newborns within 72 hours after birth remains a clinical challenge with

high morbidity and mortality [1-3]. The majority of global neonatal deaths due to EOS occur in

developing countries [4]. The diagnosis of EOS is complicated, resulting in late recognition or

overtreatment of newborns with antibiotics. These dilemmas arise because the pathophysiology

of EOS is poorly understood.

A hallmark of sepsis pathophysiology is endothelial cell activation followed by leukocyte

recruitment into tissues [5]. The current model describes the occurrence of a shift in balance

in Tie2 receptor ligands Angiopoietin (Ang)-1 and Ang-2 affecting endothelial integrity, and

increased expression of endothelial cell adhesion molecules (CAMs), in particular P-selectin,

E-selectin, vascular cell adhesion molecule (VCAM-1), and intercellular adhesion molecule

(ICAM-1) to facilitate this recruitment [6,7]. These endothelial cell adhesion molecules orchestrate

leukocyte rolling on, adhesion to, and diapedesis across the endothelium (Figure 1A) [7,8]. Also,

platelet and endothelial cell adhesion molecule (PECAM-1), expressed at endothelial cell junctions

has a function in facilitating paracellular transmigration of leukocytes across the endothelium

[9]. After intravenous administration of endotoxin in healthy adults as a sepsis model, peak levels

of Ang-2 prelude the release of soluble isoforms of CAMs (sCAMs) into the systemic circulation

[10]. CAMs are released through ectodomain shedding by enzymes called sheddases, in

particular matrix metalloproteinase-9 (MMP-9) and neutrophil elastase, released from granules

in neutrophils (Figure 1B) [7,11]. Both MMP-9 and neutrophil elastase prepare the extracellular

matrix for transmigration of leukocytes into inflammatory sites [12]. MMP-9 activity is balanced by

sheddase antagonist tissue-inhibitor of metalloproteinases-1 (TIMP-1) [12-14].

Recently, we showed in a cohort of near term and term Surinamese newborns that a systemic

circulation dysbalance in Ang-2/Ang-1 levels is associated with blood culture positive EOS [15].

This study was undertaken to examine if this dysbalance is paralleled by increased levels of sCAMs

and sheddases in this cohort of newborns with EOS to investigate their potential as biomarkers for

EOS. We hypothesized that blood culture positive EOS is associated with higher levels of sCAMs

and sheddases.

MATERIALS AND METHODSStudy Design, Subjects and Clinical Protocol

For this study, we used a Surinamese cohort of 20 healthy newborns and 71 newborns with

suspected EOS from an earlier reported study (Supplemental Table 1, previously published) [15].

All newborns were included between April 1 2015 and May 31 2016. Included were newborns with

a gestational age equal to or above 34 weeks in whom antibiotics were started within the first 72

hours of life for suspected EOS. Written informed consent was obtained from at least one parent

for the use of residual serum and clinical information. The study protocol was made available

on clinicaltrials.gov (NCT02486783) and was approved by the Surinamese Medical Ethical Board

(VG-021-14A).

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

102

6

A. Leukocyte Adhesion Cascade

B. Adhesion Molecule Shedding

Inflamed Tissue

Rolling AdhesionLateral

Migration Diapedesis

E-selectin

ICAM-1

PECAM-1

(s)ICAM-1

MMP-9 TIMP-1

MMP-9:TIMP-1 complex

(s)E-selectin

Figure 1. Schematic representation of the leukocyte adhesion cascade and shedding of endothelial

adhesion molecules. A: During inflammation circulating neutrophils first are tethered to and role on activated

endothelium, mediated by endothelial cell adhesion molecules (CAMs) P-selectin and E-selectin binding to

their ligands on leukocytes. Leukocyte rolling drives further leukocyte activation and firm adhesion through

interactions of integrins binding to their endothelial ligands including intercellular adhesion molecule-1

(ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Leukocytes then migrate on the endothelial surface

towards cell junctions rich in ICAM-1, VCAM-1, and platelet and endothelial cell adhesion molecule-1 (PECAM-1

for diapedesis across the endothelium into underlying tissues. B: During inflammation, shedding enzymes

such as neutrophil elastase and matrix metalloproteinase-9 (MMP-9) are released from granules in neutrophils.

These shedding enzymes or ‘sheddases’ release soluble isoforms of endothelial adhesion molecules (sCAMs)

into the circulation during the various stages of the leukocyte adhesion cascade. As a result, sCAM levels rise

in peripheral blood. To reduce collateral damage to host tissues, enzymatic and proteolytic activity of MMP-9

is balanced by sheddase antagonist tissue-inhibitor of metalloproteinases-1 (TIMP-1), also released from

neutrophils, by formation of inactive MMP-9:TIMP-1 complexes. Functional implications of CAM shedding

are reviewed in references 7 and 11. For simplification only endothelial adhesion molecules (s)E-selectin, (s)

ICAM-1, and (s)PECAM-1, sheddase MMP-9, and sheddase antagonist TIMP-1 are shown.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

103

6

Controls

A

B

C

Blood Culture Positive EOS

t=0 (n=20)

t=48-72 (n=3)

t=0 (n=55)

t=48-72 (n=31)

t=0 (n=5)

t=48-72 (n=2)

Blood Culture Negative EOS

D

sPEC

AM

-1 (n

g/m

L)

E

Pt=0 = 0.08Pt=48-72 = 0.59

0

250

500

750

1000

1250

1500

sVC

AM

-1 (n

g/m

L)sI

CA

M-1

(ng/

mL)

Pt=0 = 0.84 Pt=48-72 = 0.24

sE-s

elec

tin (n

g/m

L)

Pt=0 = 0.25 Pt=48-72 = 0.53

0

100

200

300

400

0

10

20

30

40

Pt=0 = 0.35Pt=48-72 = 0.90

0

200

400

600

1000

1500

Pt=0 = 0.32 Pt=48-72 = 0.07

0

100

200

300

sP-s

elec

tin (n

g/m

L)

Figure 2. Circulating levels of endothelial adhesion molecules sP-selectin, sE-selectin, sVCAM-1,

sICAM-1, and sPECAM-1 in Surinamese newborns. A: sP-selectin B: sE-selectin C: soluble vascular cell

adhesion molecule-1 (sVCAM-1); D: soluble intercellular adhesion molecule-1 (sICAM-1); E: soluble platelet

and endothelial cell adhesion molecule-1 (sPECAM-1). Data report levels in serum sampled at t=0 (white bars)

and t=48-72h (grey bars) and are analyzed with a Kruskal-Wallis test between all groups at t=0 (Pt=0

) and at

t=48-72 (Pt=48-72

). P<0.05 is considered statistically significant. Bars represent median values and error bars

interquartile range.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

104

6

Controls

A

B

C

Blood Culture Positive EOS

t=0 (n=20)

t=48-72 (n=3)

Pt=0 = 0.84Pt=48-72 = 0.35

t=0 (n=64)

t=48-72 (n=44)

t=0 (n=6)

t=48-72 (n=3)

Blood Culture Negative EOS

MM

P-9

(ng/

mL)

Pt=0 = 0.13Pt=48-72 = 0.23

TIM

P-1/

MM

P-9

Pt=0 = 0.90Pt=48-72 = 0.43

0

250

500

750

1000

1250

1500

0

250

500

750

1000

TIM

P-1

(ng/

mL)

0

1

2

3

4

5

6

Figure 3. Circulating levels of MMP-9 and TIMP-1, and TIMP-1/MMP-9 ratios in Surinamese newborns.

A: Matrix metalloproteinase-9 (MMP-9); B: Tissue inhibitor of metalloproteinase (TIMP-1); C: TIMP-1/MMP-9

ratios. Data report levels in serum sampled at t=0 (white bars) and t=48-72h (grey bars) and are analyzed with

a Kruskal-Wallis test between all groups at t=0 (Pt=0

) and at t=48-72 (Pt=48-72

). P<0.05 is considered statistically

significant. Bars represent median values and error bars interquartile range.

The management of these patients was described before [15]. In short, healthy control

newborns and newborns suspected of EOS were included (t=0) within 72 hours after birth and

clinically reevaluated 48-72 hours later (t=48-72h). At t=0 and t=48-72h blood was drawn for

separation and storage of serum. Controls were newborns without signs of infection receiving

blood draws for hyperbilirubinemia (n=20). Newborns with suspected EOS receiving treatment

with intravenous antibiotics were divided in two groups based on result from blood culturing:

blood culture negative EOS (n=65) and blood culture positive EOS (n=6).

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

105

6

Sample Collection, Preparation and Analysis

At t=0 blood samples were collected during the insertion of a venous cannula and after 48-72

hours of treatment with antibiotics a second blood sample was obtained using capillary collection.

After clotting at room temperature and centrifugation at 2,300xg for 8 minutes the serum was

harvested and the residual sample was stored at -80°C until further analysis. Measurement of

sP-selectin, sE-selectin, sVCAM-1, sICAM-1, and sPECAM-1 was performed on serum samples

using the Human Magnetic Bead Adhesion 6-plex panel performance assay (LHC0016M, Thermo

Scientific, Waltham, MA USA) according to the manufacturer’s instructions. ELISA was used on

aliquots of the same samples for measurement of neutrophil elastase (HK319-02, Hycult Biotech,

Uden, The Netherlands), MMP-9 (Quantikine DMP900, R&D systems, Minneapolis, MN USA), and

TIMP-1 (Quantikine DTM100, R&D systems), each according to the manufacturers’ instructions.

Serum samples (n=142) were available of all 91 newborns at t=0 and of 51 at t=48-72h. Due

to the limited amount of serum available, not all molecules could be measured in all samples.

Measurement of levels of MMP-9 and TIMP-1 was performed in n=90 and n=51 of newborns at

t=0 and t=48-72h, respectively. We were able to measure sCAMs and neutrophil elastase levels in

n=80 and n=36 newborns at t=0 and 48-72h, respectively. For each molecule, a standard curve was

established via which concentrations in neonatal serum were determined. Levels below or above

the linear part (for MMP-9 n=11 (7.7%) samples, for TIMP-1 n=2 (1.4%) samples, and for neutrophil

elastase n=9 (6.3%) samples) of this standard curve were reported as the lowest or highest value

of the standard curve, respectively. We measured intra-assay variation between plates used in

the same assay by calculating coefficient of variation between levels of each molecule in samples

from the same patient divided over those plates and accepted a maximum of 20%.

Controls Blood Culture Positive EOS

t=0 (n=20)

t=48-72 (n=3)

t=0 (n=55)

t=48-72 (n=31)

t=0 (n=5)

t=48-72 (n=2)

Blood Culture Negative EOS

0

1000

2000

3000

4000

Neu

troph

il El

asta

se (n

g/m

L)

Pt=0 = 0.06Pt=48-72 = 0.31

Figure 4. Circulating levels of neutrophil elastase in Surinamese newborns. Data report levels in serum

sampled at t=0 (white bars) and t=48-72h (grey bars) and are analyzed with a Kruskal-Wallis test between all

groups at t=0 (Pt=0

) and at t=48-72 (Pt=48-72

). P<0.05 is considered statistically significant. Bars represent median

values and error bars interquartile range.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

106

6

Statistical Analysis

Categorical variables were presented as numbers and percentages with 95% CI and continuous

variables, due to the nonparametric nature of the data, as median with interquartile range (IQR).

The Chi-square test was used to compare categorical variables. A Mann-Whitney U test and Kruskal-

Wallis test with Dunn’s correction for multiple comparisons were used for analysis of continuous

variables. Because timing of inclusion after birth varied between groups (Supplemental Table 1),

we investigated whether postnatal sampling day (i.e., for both t=0 and t=48-72h between day 1 and

6 after birth) correlated with sCAM and sheddase levels and calculated Spearman’s rho. P-values

<0.05 were considered statistically significant. All analyses were done using Prism version 7.0a

(Graphpad Software Inc., San Diego, CA USA).

RESULTSDemographic variables of the study cohort (n=91) are given in Supplemental Table 1. Blood culture

results revealed that 6 of 71 newborns with suspected EOS (8.5%; 95% CI 3.9-17.2) had a positive

blood culture with gram-negative pathogens Klebsiella pneumoniae (n=2), Enterobacter cloacae

(n=2) and Escherichia coli (n=2). One newborn had EOS due to a spontaneous bacterial peritonitis.

For n=4 others cause of EOS was unknown, but they presented with neonatal jaundice (n=1),

perinatal asphyxia (n=1), meconium aspiration (n=1), and hypoglycaemia (n=1).

Serum Levels of Endothelial Cell Adhesion Molecules

At t=0, no differences between blood culture positive EOS and controls were found for median

levels of sP-selectin (169 (99.9) ng/mL versus 172 (98) ng/mL, respectively; P=0.41), sE-selectin (401

(1067) ng/mL versus 360 (639) ng/mL, respectively; P=0.24), sVCAM-1 (1134 (134) ng/mL versus 1170

(46) ng/mL, respectively; P=0.49), sICAM-1 (135 (215) ng/mL versus 124 (123) ng/mL, respectively;

P=0.83), sPECAM-1 (18 (14) ng/mL versus 20 (8) ng/mL, respectively; P=0.97). No differences in

sCAM levels between t=0 and t=48-72h were found with a Mann-Whitney U test. No differences

between median levels of sCAMs between controls, blood culture negative EOS and blood culture

positive EOS groups at either t=0 or t=48-72h were found with a Kruskal-Wallis test (Figure 2A-E).

Of all sCAMs only median levels of sP-selectin in pooled (n=115) samples correlated negatively with

later sampling day (rho -0.21; 95% CI -0.38 to -0.02; P=0.03).

Serum Levels of MMP-9, TIMP-1, and Neutrophil Elastase

At t=0, median levels of sheddase MMP-9 (462 (599) ng/mL versus 420 (1027) ng/mL, respectively;

P>0.99), TIMP-1 (447 (300) ng/mL versus 288 (80) ng/mL, respectively; P=0.14) and TIMP/MMP-9

ratios (0.7 (4.1) versus 0.7 (1.7), respectively; P=0.66) were not different between blood culture

positive EOS and controls. Neutrophil elastase levels were similar between blood culture positive

EOS and controls (1201 (2566) ng/mL versus 1081 (808) ng/mL, respectively; P=0.57). For none of

the molecules median levels were different at t=48-72h from t=0. No differences between median

levels of sheddases between controls, blood culture negative EOS, and blood culture positive

EOS at either t=0 or t=48-72h was found with a Kruskal-Wallis test (Figure 3A-C and Figure 4). No

correlation was found between levels of sheddases and sampling day.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

107

6

DISCUSSIONIn this study we investigated whether sCAMs and their sheddases circulate at higher levels in

newborns with blood culture positive EOS. We serially measured levels of sCAMs and sheddases

in a cohort of near and at term newborns. In contrast to our hypothesis, none of the molecules

showed any difference in serum levels between blood culture positive EOS and controls, neither

at start of antibiotic treatment nor after 48-72 hours. These data indicate that levels of sCAM and

sheddases are of limited clinical utility as early biomarkers for EOS in newborns.

Previously, we found evidence for endothelial cell activation in blood culture positive EOS in

the same newborns used for this study, represented by a dysbalance in Ang-2/Ang-1 ratio [15].

Since the current data demonstrate that this dysbalance was not paralleled by increased release of

sCAM or sheddases in EOS we conclude that CAM shedding is not or to a lesser extent involved in

the pathophysiology of EOS. For interpretation of our data we reviewed and summarized available

data on sCAMs and sheddases in newborns with sepsis in Supplemental Table 2. Comparison of our

results with other existing data is complicated because of heterogenic make up of chosen cohorts.

Only one study reported a comparable cohort of near and at term newborns with suspected EOS

within 72 hours after birth, in whom increased levels of sICAM-1 and neutrophil elastase levels were

associated with blood culture positive EOS [19]. Other earlier studies compared levels of sCAMs

in heterogenic cohorts consisting of newborns with different gestational and postnatal ages,

either having EOS (based on varying definitions), or sepsis after 72 hours after birth (i.e., late onset

sepsis). This variation in inclusion criteria is an important confounding factor in the interpretation

of the observed levels in septic and healthy newborns. Overall, our results are in line with these

studies that show that clinical utility of levels of sCAMs and sheddases in EOS is limited.

In an earlier review by our group we pooled published data on sCAM levels in newborns [7].

Soluble CAM levels in the current study corresponded well with levels discussed in our review

and those established in earlier studies in uninfected healthy newborns with similar gestational

and postnatal age [7,23,24,29-32]. However, MMP-9, TIMP-1, and neutrophil elastase levels were

different and up to 4, 2, and 10-fold higher, respectively, than those reported in earlier studies

[17,18,21,22,25,30], which may have been due to other methods used (see limitations). Furthermore,

our earlier review and earlier data indicated that significant age-related discrepancies exist in

sCAM levels between newborns, children and adults. As an example, in at term newborns sVCAM-1

concentrations in the first postnatal week were almost twice the levels in healthy adults, and equally

high compared to septic adults, suggesting that sVCAM-1 levels start of high in early newborn life

and then decrease with increasing age [7,31]. In our study, levels of sCAMs and sheddases during

the first 6 days of life in our study remained stable for 7 out of analyzed 8 molecules, which was in

contrast with earlier work in healthy newborns showing that sE-selectin decreased, and sICAM-1

and sVCAM-1 increased between day 1 and 5 after birth, while sPECAM-1 levels did not change

[29-32]. Even though some discrepancies with earlier reports exist, overall one can conclude that

these and our data indicate that levels of sCAMs and sheddases measured within 72 hours after

birth are high and do not discriminate between septic and healthy newborns, which limits their

use as biomarkers for early identification or exclusion of EOS.

Our and pre-existing data suggest that overall high sCAM and sheddase levels in newborns are

the result of other perinatal factors than EOS. Several pathophysiological processes may explain

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

108

6

this premise. Birth may induce a ‘pro-adhesive’ state of the endothelium leading to increased CAM

expression on, and CAM shedding from, its surface. Additionally, the increase in overall leukocyte

numbers and inflammatory activation of subsets associated with human birth, which was shown

to be positively associated with increased perinatal stress [35-37], may cause higher intensity of

leukocyte-endothelial interactions and subsequent increases CAM shedding. Aberrant adhesion

of activated leukocytes to activated endothelium is associated with endothelial dysfunction

and increased vascular permeability [38,39]. Shedding of CAMs may then result in prevention of

aberrant leukocyte adhesion on two complementary levels, namely 1) to lower endothelial CAM

density to prevent adhesion or promote de-adhesion of already adhering leukocytes and 2) to

release circulating sCAMs that act as ‘decoy receptors’ to capture leukocytes in the vasculature

to limit leukocyte-endothelial interactions [7,10]. Whether this occurs in real life and what

the contribution is to sCAM and sheddase levels in newborns remains unknown and could be

studied in neonatal animal models [40-42].

Our study has some limitations. First, sample size at t=48-72h was relatively small due to

limited clinical need for additional blood draws in controls and death of patients. As a result,

logistic regression analysis of other factors, such as maternal perinatal factors or method of birth,

potentially influencing levels of sCAMs and sheddases, was precluded. Larger studies in countries

such as Suriname, where we expect incidence of EOS to be relatively high in comparison to Western

countries, are necessary and can contribute to better insight in the vascular pathophysiology of

EOS. Second, the use of serum in our study may have caused release of stored pools of MMP-9,

TIMP-1, and neutrophil elastase from disrupted leukocytes during the clotting process, which

could have accounted for higher levels of these molecules than reported in earlier studies.

In conclusion, our data indicate that serum levels of sCAMs and sheddases are not increased

during EOS in Surinamese near and at term newborns. Other mechanisms, such as perinatal stress

during birth, may drive overall high levels in all newborns which precludes discrimination between

septic and healthy newborns based on levels of these molecules. For these reasons sCAMs and

sheddases studied have limited utility as biomarkers for EOS.

Abbreviations and Definitions

EOS = Early onset sepsis

ICAM-1 = Intercellular adhesion molecule-1

VCAM-1= Vascular cell adhesion molecule-1

PECAM-1= Platelet and endothelial cell adhesion molecule-1

MMP-9 = Matrix metalloproteinase-9

TIMP-1 = Tissue-inhibitor of metalloproteinases-1

Funding

The research in this study was supported by the Thrasher Research Fund (TRF13064) (R. Zonneveld)

and Tergooi Hospitals, Blaricum, The Netherlands.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

109

6

Acknowledgments

The authors acknowledge the efforts of all employees of the Clinical Laboratory of the Academic

Hospital Paramaribo and the Central Laboratory of Suriname, Paramaribo, Suriname, for assistance

with sample storage, handling and transport.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

110

6

REFERENCES1. Van Herk W, Stocker M, van Rossum AM.

Recognising early onset neonatal sepsis: an

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77-82.

2. Simonson KA, Anderson-Berry AL, Delair SF,

Davies HD. Early-onset neonatal sepsis. Clin

Microbiol Rev 2014, 27(1):21-47.

3. Stoll BJ. Early-onset neonatal sepsis: a continuing

problem in need of novel prevention strategies.

Pediatrics 2016, 138(6): e20163038.

4. Lawn JE, Cousens S, Zupan J; Lancet

Neonatal Survival Steering Team. 4 million

neonatal deaths: when? Where? Why?

Lancet 2005, 365(9462):891-900.

5. Aird WC. The role of the endothelium in

severe sepsis and multiple organ dysfunction

syndrome. Blood 2003, 101(10):3765-77.

6. Fiedler U, Augustin HG. Angiopoietins:

a link between angiogenesis and inflammation

Trends Immunol 2006, 27:552-558.

7. Zonneveld R, Martinelli R, Zonneveld R,

Martinelli R, Shapiro NI, Kuijpers TW, Plötz

FB, Carman CV. Soluble adhesion molecules

as markers for sepsis and the potential

pathophysiological discrepancy in neonates,

children and adults. Crit Care 2014, 18;18(1):204.

8. Ley K, Laudanna C, Cybulsky MI, Nourshargh

S. Getting to the site of inflammation:

the leukocyte adhesion cascade updated. Nat

Rev Immunol 2007, 7(9):678-89.

9. Privratsky JR, Newman PJ. PECAM-1: regulator

of endothelial junctional integrity. Cell Tissue

Res 2014, 355(3): 607–619.

10. Kümpers P, van Meurs M, David S, Molema G,

Bijzet J, Lukasz A, Biertz F, Haller H, Zijlstra JG.

Time course of angiopoietin-2 release during

experimental human endotoxemia and sepsis.

Crit Care 2009, 13(3):R64.

11. Garton KJ, Gough PJ, Raines EW. Emerging roles

for ectodomain shedding in the regulation

of inflammatory responses. J Leukoc

Biol 2006, 79(6):1105-16.

12. Elkington PTG, O’Kane CM, Friedland JS.

The paradox of matrix metalloproteinases

in infectious disease. Clin Exp

Immunol 2005, 142(1): 12-20.

13. Brew K, Nagase H. The tissue inhibitors of

metalloproteinases (TIMPs): An ancient

family with structural and functional diversity.

Biochim Biophys Acta 2010, 1803(1): 55–71.

14. Lorente L, Martín MM, Solé-Violán J, Blanquer

J, Labarta L, Díaz C, Borreguero-León JM,

Orbe J, Rodríguez JA, Jiménez A, Páramo JA.

Association of sepsis-related mortality with

early increase of TIMP-1/MMP-9 ratio. PLoS

One 2014, 11;9(4):e94318.

15. Zonneveld R, Jongman R, Juliana A, Zijlmans W,

Plötz FB, Molema G, Van Meurs M. Low Serum

Angiopoietin-1, high Angiopoietin-2, and high

Ang-2/Ang-1 Protein Ratio are Associated with

Early Onset Sepsis in Surinamese Newborns.

Shock 2017, in press.

16. Fattah MA, Omer AF, Asaif S, Manlulu R, Karar T,

Ahmed A, Aljada A, Saleh AM, Qureshi S, Nasr A.

Utility of cytokine, adhesion molecule and acute

phase proteins in early diagnosis of neonatal

sepsis. J Nat Sci Biol Med 2017, 8(1):32-39.

17. Weitkamp JH, Guthrie SO, Wong HR,

Moldawer LL, Baker HV, Wynn JL. Histological

chorioamnionitis shapes the neonatal

transcriptomic immune response. Early Hum

Dev 2016, 98:1-6.

18. Wynn JL, Guthrie SO, Wong HR, Lahni P,

Ungaro R, Lopez MC, Baker HV, Moldawer

LL. Postnatal Age Is a Critical Determinant of

the Neonatal Host Response to Sepsis. Mol

Med. 2015;21:496–504

19. Sugitharini V, Prema A, Berla Thangam

E. Inflammatory mediators of systemic

inflammation in neonatal sepsis. Inflamm

Res 2013, 62(12):1025-34.

20. Edgar JDM, Gabriel V, Gallimore JR,

McMillan SA, Grant J. A prospective study

of the sensitivity, specificity and diagnostic

performance of soluble intercellular adhesion

molecule 1, highly sensitive C-reactive

protein, soluble E-selectin and serum amyloid

A in the diagnosis of neonatal infection. BMC

Pediatr 2010, 16:10–22.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

111

6

21. Fukunaga S, Ichiyama T, Maeba S, Okuda M,

Nakata M, Sugino N, Furukawa S. MMP-9 and

TIMP-1 in the cord blood of premature infants

developing BPD. Ped Pulm 2009, 44: 267-272.

22. Sunagawa S, Ichiyama T, Honda R,

Fukunaga S, Maeba S, Furukawa S. Matrix

metalloproteinase-9 and tissue inhibitor of

metalloproteinase-1 in perinatal asphyxia.

Brain Dev 2009, 31(8):588-93.

23. Figueras-Aloy J, Gómez-López L, Rodríguez-

Miguélez JM, Salvia-Roiges MD, Jordán-

García I, Ferrer-Codina I, Carbonell-Estrany X,

Jiménez-González R. Serum soluble ICAM-1,

VCAM-1, L-selectin and P-selectin levels

as markers of infection and their relation

to clinical severity in neonatal sepsis. Am J

Perinat 2007, 24:331–338.

24. Sitaru AG, Speer CP, Holzhauer S, Obergfell

A, Walter U, Grossmann R. Chorioamnionitis

is associated with increased CD40L

expression on cord blood platelets. Thromb

Haemost 2005, 94(6):1219-23.

25. Schulz CG, Sawicki G, Lemke RP, Roeten BM,

Schulz R, Cheung PY. MMP-2 and MMP-9

and their tissue inhibitors in the plasma

of preterm and term neonates. Pediatr

Res 2004, 55(5):794-801.

26. Edgar JDM, Gabriel V, Craig A, Wheeler D,

Thomas M, Grant J. A low serum sICAM-1

level may assist in the exclusion of neonatal

infection. Biol Neonate 2002, 81:105–108.

27. Apostolou M, Dimitriou H, Kaleyias J,

Perdikogianni C, Stiakaki E, Costalos C,

Kalmanti MC. Levels of soluble ICAM-1 in

premature and full-term neonates with

infection. Mediators Inflamm 2002, 11:95–98.

28. Dollner H, Vatten L, Austgulen R. Early diagnostic

markers for neonatal sepsis: comparing

C-reactive protein, interleukin-6, soluble tumor

necrosis factor receptors and soluble adhesion

molecules. J Clin Epidemiol 2001, 54:1251–1257.

29. Malamitsi-Puchner A, Tziotis J, Mastorakos

G, Protonotariou E, Creatsas G. Adhesion

molecules in early neonatal life. Biol

Neonate 2000, 78(1):65-7.

30. Giannaki G, Rizos D, Xyni K, Sarandakou A,

Protonotariou E, Phocas I, Creatsas G. Serum

soluble E-and L-selectin in the very early neonatal

period. Early Hum Develop 2000, 60:149–155.

31. Giannaki G, Xyni K, Rizos D, Phocas I.

Comparative study of serum soluble VCAM-1

and ICAM-1 levels in the early neonatal period.

Acta Paediatr 1999, 88(12):1413-4.

32. Phocas I, Sarandakou A, Giannaki G, Malamitsi-

Puchner A, Rizos D, Zourlas PA. Soluble

intercellular adhesion molecule-1 in newborn

infants. Eur J Pediatr 1998, 157(2):153-6.

33. Berner R, Niemeyer CM, Leititis JU, Funke A,

Schwab C, Rau U, Richter K, Tawfeek MS, Clad A,

Brandis M. Plasma levels and gene expression

of granulocyte colony-stimulating factor,

tumor necrosis factor-(alpha), interleukin

(IL)-1(beta), IL-6, IL-8, and soluble intercellular

adhesion molecule-1 in neonatal early onset

sepsis. Pediatr Res 1998, 44:469–477.

34. Austgulen R, Arntzen KJ, Haereid PE, Aag S,

Døllner H. Infections in neonates delivered

at term are associated with increased

serum levels of ICAM-1 and E-selectin. Acta

Pediatr 1997, 86:274–280.

35. Lim FT, van Winsen L, Willemze R, Kanhai

HH, Falkenburg JH. Influence of delivery

on numbers of leukocytes, leukocyte

subpopulations, and hematopoietic

progenitor cells in human umbilical cord

blood. Blood Cells 1994,20(2-3):547-58;

discussion 558-9.

36. Lim FT, Scherjon SA, van Beckhoven JM, Brand

A, Kanhai HH, Hermans JM, Falkenburg JH.

Association of stress during delivery with

increased numbers of nucleated cells and

hematopoietic progenitor cells in umbilical cord

blood. Am J Obstet Gynecol 2000, 183(5):1144-52

37. Duijts L, Bakker-Jonges LE, Labout JA, Jaddoe VW,

Hofman A, Steegers EA, Van Dongen JJ, Hooijkaas

H, Moll HA. Perinatal stress influences lymphocyte

subset counts in neonates. The generation R

study. Pediatr Res 2008, 63(3):292-8.

38. Beyrau M, Bodkin JV, Nourshargh S.

Neutrophil heterogeneity in health and

disease: a revitalized avenue in inflammation

and immunity. Open Biol 2012, 2(11):120134.

39. Brown KA, Brain SD, Pearson JD, Edgeworth

JD, Lewis SM, Treacher DF. Neutrophils in

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

112

6

development of multiple organ failure in

sepsis. Lancet 2006, 368(9530):157-69.

40. Shapiro NI, Yano K, Sorasaki M, Fischer

C, Shih SC, Aird WC. Skin biopsies

demonstrate site-specific endothelial

activation in mouse models of sepsis. J Vasc

Res 2009, 46(5):495-502.

41. Csonka K, Vadovics M, Marton A, Vágvölgyi C,

Zajta E, Tóth A, Tóth R, Vizler C, Tiszlavicz L,

Mora-Montes HM, Gácser A. Investigation of

OCH1 in the Virulence of Candida parapsilosis

Using a New Neonatal Mouse Model. Front

Microbiol 2017, 8:1197.

42. Molina-Berríos A, Campos-Estrada C, Lapier

M, Duaso J, Kemmerling U, Galanti N, Ferreira

J, Morello A, López-Muñoz R, Maya JD.

Protection of vascular endothelium by aspirin

in a murine model of chronic Chagas’ disease.

Parasitol Res 2013, 112(7):2731-9.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

113

6

SUPPLEMENTAL DATA

Supplemental Table 1. Descriptive statistics of the study group (n=91)

Controls

(n=20)

Early Onset Sepsis

P-value

Blood Culture

Negative (n=65)

Blood Culture

Positive (n=6)

Pregnancy, n (%) Complications1

Chorioamnionitis2

3 (15)

0

16 (25)

18 (28)

1 (17)

0

0.63

Mode of delivery,

n (%)

Vaginal

Caesarean

12 (60)

8 (40)

46 (75)

19 (25)

4 (67)

2 (33)

0.54

Sex, n (%) Male

Female

9 (45)

11 (55)

29 (45)

36 (55)

5 (83)

1 (17)

0.19

Ethnicity, n (%) Maroon and Creole

Hindo-Surinamese

Other3

12 (60)

3 (15)

5 (25)

44 (68)

14 (21)

7 (11)

4 (67)

1 (17)

1 (17)

0.61

Gestational age,

n (%) (weeks)

34-37

37-40

≥40

1 (5)

14 (70)

5 (25)

22 (34)

30 (46)

13 (20)

0

4 (67)

2 (33)

0.06

Apgar score, n (%) <5 0 5 (8) 2 (33) 0.03

Birth weight, Median

(IQR) (grams)

3130 (700) 2840 (835) 3500 (906) 0.02

Age at presentation,

n (%) (hours)

<24

24-48

48-72

4 (20)

7 (35)

9 (45)

43 (66)

13 (20)

9 (14)

2 (33)

1 (17)

3 (50)

<0.01

Clinical course

(at 48-72h), n (%)

CPAP

Mechanical Ventilation

Cardiotonics

Mortality

0

0

0

0

9 (14)

7 (11)

5 (8)

3 (5)

0

2 (33)

1 (17)

2 (33)

<0.001

CPAP = continuous positive airway pressure; N/A = not applicable. 1 Presence of pregnancy-induced hypertension, preeclampsia or diabetes mellitus.2 Defined as intrapartum fever or administration of antibiotics.3 Includes: Javanese, Chinese, Caucasian and Amerindian.

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

114

6

Sup

ple

men

tal T

able

2. S

tudi

es r

epo

rtin

g le

vels

of e

ndo

thel

ial c

ell a

dhes

ion

mo

lecu

les

and

shed

ding

enz

ymes

in n

ewb

orn

s

Stud

y [r

ef.]

, yea

r

CA

M o

r

shed

din

g e

nzy

me

Co

ho

rt

Ch

arac

teri

stic

s

Ges

tati

on

al

age

(wee

ks)

Post

nat

al

age

Mai

n R

esul

ts

An

alys

is

Met

ho

d

Hea

lth

y

(ng

/mL)

1

Sep

tic

(ng

/mL)

1

Fata

h et

al.

[16]

,

2017

sE-s

elec

tin

EOS

and

LO

SN

SN

SsE

-sel

ecti

n el

evat

ed in

BC

PSEL

ISA

148.

9 ±

7.917

7.1 ±

3.5

Wei

tkam

p et

al.

[17]

, 20

16

MM

P-9

EOS

and

LO

S25

-36

< an

d ≥

3

day

s

MM

P-9

leve

ls lo

wer

in B

CPS

M

ulti

plex

bea

d

assa

y

NS

NS

Wyn

n et

al.

[18]

,

2015

MM

P-9

Cho

rio

amni

oni

tis

25-3

6N

SM

MP-

9 le

vels

low

er in

cho

rio

amni

oni

tis

Mul

tipl

ex b

ead

assa

y

NS

NS

Sugi

thar

ini e

t al

.

[19]

, 20

13

sIC

AM

-1, N

EEO

S34

-42

0-7

2 ho

urs

sIC

AM

-1 a

nd N

E el

evat

ed

in E

OS

ELIS

A

Ant

ibo

dy

arra

y

sIC

AM

-1/

NE:

NS

sIC

AM

-1: N

S

NE:

499.

2±22

.0

Edga

r et

al.

[20

],

2010

sIC

AM

-1, s

E-se

lect

inEO

S an

d L

OS

24-4

1N

SsI

CA

M-1

and

sE-

sele

ctin

elev

ated

ELIS

AsI

CA

M-1

: 165

(130

-290

)

sE-s

elec

tin:

71

(51-

118)

sIC

AM

-1: 4

05

(252

-666

)

sE-s

elec

tin

158

(94-

207)

Fuka

naga

et

al.

[21]

, 20

09

MM

P, T

IMP-

1U

ninf

ecte

d

new

bo

rns

<30

Co

rd

blo

od

No

diff

eren

ceEL

ISA

MM

P-9:

22

(16-

48)

TIM

P-1:

122

(86-

249)

NS

Suna

gaw

a et

al.

[22]

, 20

09

MM

P-9,

TIM

P-1

Uni

nfec

ted

new

bo

rns

35-4

11-

2 d

ays

NA

ELIS

AN

SN

A

Figu

eras

et

al.

[23]

, 20

07

sIC

AM

-1, s

VC

AM

-1,

sP-s

elec

tin,

EOS

and

LO

S32

-40

1-32

day

ssI

CA

M-1

and

sV

CA

M-1

incr

ease

d o

ver

tim

e.

ELIS

AsI

CA

M-1

: 156

(150

-194

)

sVC

AM

-1: 8

56

(742

-960

)

sP-s

elec

tin:

272

(152

-288

)

sIC

AM

-1: 3

94

(342

-60

0)

sVC

AM

-1: 1

153

(726

-130

7)

sP-s

elec

tin:

244

(170

-324

)

Sita

ru e

t al

. [24

],

200

5

sP-s

elec

tin

Cho

rio

amni

oni

tis

25-4

0C

ord

blo

od

sP-s

elec

tin

elev

ated

in

cho

rio

amni

oni

tis

ELIS

A10

4 ±

7122

2 ±

128

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

115

6

Sup

ple

men

tal T

able

2. (

cont

inue

d)

Stud

y [r

ef.]

, yea

r

CA

M o

r

shed

din

g e

nzy

me

Co

ho

rt

Ch

arac

teri

stic

s

Ges

tati

on

al

age

(wee

ks)

Post

nat

al

age

Mai

n R

esul

ts

An

alys

is

Met

ho

d

Hea

lth

y

(ng

/mL)

1

Sep

tic

(ng

/mL)

1

Schu

lz e

t al

. [25

],

200

4

MM

P-9,

TIM

P-1

Uni

nfec

ted

new

bo

rns

25-4

01-

28 d

ays

MM

P-9

high

est

in p

rete

rm

TIM

P-1 h

ighe

st in

at

term

ELIS

AN

SN

A

Edga

r et

al.

[26]

,

200

2

sIC

AM

-1EO

S an

d L

OS

24-4

2N

SsI

CA

M-1

ele

vate

d in

BC

PSEL

ISA

205

(146

-343

)40

6 (3

45-1

180

)

Ap

ost

olo

u et

al.

[27]

, 20

02

sIC

AM

-1EO

S an

d L

OS

25-4

2N

SsI

CA

M-1

ele

vate

d in

BC

PS

ELIS

A35

8.4

± 28

.971

0.7

± 5

6.6

Do

llner

et

al. [

28],

200

1

sIC

AM

-1, s

E-se

lect

inEO

S an

d L

OS

30-4

21-

7 d

ays

sIC

AM

-1 a

nd s

E-se

lect

in

elev

ated

in B

CPS

ELIS

AsI

CA

M-1

: 244

.0

(92.

5-50

0)

sE-s

elec

tin:

91.4

(<2.

0-2

17.8

)

sIC

AM

-1: 3

57.4

(141

.6-5

00

)

sE-s

elec

tin:

151.

7 (3

7.0

-

362.

2)

Mal

amit

si e

t al

.

[29]

, 20

00

sVC

AM

-1, s

PEC

AM

-1U

ninf

ecte

d

new

bo

rns

37-4

01-

5 d

ays

No

cha

nge

bet

wee

n d

ay 1

and

5

ELIS

AsV

CA

M-1

: 134

0

± 58

.3

sPEC

AM

-1: 1

7.5

± 0

.7

NA

Gia

nnak

i et

al.

[30

], 2

00

0

sE-s

elec

tin

Uni

nfec

ted

new

bo

rns

At

term

1-5

day

ssE

-sel

ecti

n d

ecre

ases

bet

wee

n d

ay 1

and

5

ELIS

A13

9 ±

48N

A

Gia

nnak

i et

al.

[31]

, 199

9

sIC

AM

-1, s

VC

AM

-1U

ninf

ecte

d

new

bo

rns

At

term

1-5

day

ssI

CA

M-1

and

sV

CA

M-1

incr

ease

bet

wee

n d

ay 1

and

5

ELIS

AsI

CA

M: 1

79 ±

56.1

sVC

AM

-1: 1

125.

0

± 28

1.0

NA

Pho

cas

et a

l. [3

2],

1998

sIC

AM

-1U

ninf

ecte

d

new

bo

rns

35-4

21-

30 d

ays

sIC

AM

-1 in

crea

ses

bet

wee

n

day

1, 5

and

30

ELIS

A13

7.3

± 62

.0N

A

Bern

er e

t al

. [33

],

1998

sIC

AM

-1EO

S26

-42

0-9

6

hour

s

sIC

AM

-1 lo

wer

in E

OS.

sIC

AM

-1 in

crea

ses

ove

r ti

me

ELIS

A42

1 (29

1-45

9)44

6 (1

71-5

34)

AD

HESIO

N M

OLEC

ULE SH

EDD

ING

IN SU

RINA

MESE N

EWBO

RNS

116

6

Sup

ple

men

tal T

able

2. (

cont

inue

d)

Stud

y [r

ef.]

, yea

r

CA

M o

r

shed

din

g e

nzy

me

Co

ho

rt

Ch

arac

teri

stic

s

Ges

tati

on

al

age

(wee

ks)

Post

nat

al

age

Mai

n R

esul

ts

An

alys

is

Met

ho

d

Hea

lth

y

(ng

/mL)

1

Sep

tic

(ng

/mL)

1

Aus

tgul

en e

t al

.

[34]

, 199

7

sIC

AM

-1, s

VC

AM

-1,

sE-s

elec

tin

EOS

and

LO

S,

pneu

mo

nia

24-4

20

-162

hour

s

sE-s

elec

tin

and

sIC

AM

-1

elev

ated

in in

fect

ed

neo

nate

s

ELIS

AsE

-sel

ecti

n: 8

4.2

(21.

6-23

1.3)

sIC

AM

-1: 2

131.

3

(144

9.5-

350

0.0

)

sVC

AM

-1: 2

37.0

(122

.0-5

00

.0)

NS

CA

M =

end

oth

elia

l ce

ll ad

hesi

on

mo

lecu

le;

sVC

AM

-1 =

so

lubl

e V

ascu

lar

Cel

l A

dhe

sio

n M

ole

cule

-1;

sIC

AM

-1 =

so

lubl

e In

terc

ellu

lar

Ad

hesi

on

Mo

lecu

le-1

; N

E =

neut

roph

il el

asta

se N

A =

No

t

avai

labl

e; N

S =

No

t sp

ecifi

ed; E

OS

= Ea

rly

Ons

et S

epsi

s; L

OS

= La

te O

nset

Sep

sis;

BC

PS =

Blo

od

Cul

ture

Po

siti

ve S

epsi

s.a Le

vels

are

in m

ean

± SD

, mea

n ±

SEM

, med

ian

(int

erq

uart

ile r

ange

), o

r m

edia

n (r

ange

).

7 Low Serum Angiopoietin-1, high Angiopoietin-2, and high Ang-2/Ang-1

Protein Ratio are Associated with Early Onset Sepsis in Surinamese Newborns

Rens Zonneveld, Rianne M. Jongman, Amadu Juliana, Wilco Zijlmans, Frans B. Plötz,

Grietje Molema, Matijs van Meurs

Shock 2017, May 22

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

120

7

ABSTRACTPurpose

Vascular inflammation and leakage in sepsis is mediated by Angiopoietin-1 (Ang-1) and -2 (Ang-2)

and their phosphorylation of the endothelial Tie-2 receptor. Levels of Ang-2 change in adults and

children during sepsis, which is associated with severity of sepsis. This study investigates levels of

Ang-1 and Ang-2 in newborns to gain insight in the vascular pathophysiology of early onset sepsis

(EOS) within 72 hours after birth.

Methods

A prospective cohort study was performed amongst 71 Surinamese newborns treated with

antibiotics for suspected EOS and 20 control newborns. Newborns with suspected EOS were

divided in two groups: blood culture negative and positive EOS. Ang-1 and Ang-2 levels were

measured in serum obtained at start of antibiotic treatment and at reevaluation after 48-72 hours.

Results

In this cohort 8.5% of newborns had a positive blood culture. At start of antibiotic treatment

Ang-1 serum levels were lower (P<0.01), and Ang-2 and Ang-2/Ang-1 serum protein ratios higher

(P<0.01 and P<0.01, respectively) in newborns with blood culture positive EOS than in controls.

These levels were not dependent on timing of first blood draw after birth. After 48-72 hours levels

of Ang-1 further decreased in blood culture positive EOS, while in the other groups no change

was observed.

Conclusions

Our findings support the hypothesis that a dysbalance in the Angiopoietins plays a role in

the vascular pathophysiology of EOS.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

121

7

INTRODUCTIONSepsis is a syndrome with physiologic, pathological and biochemical changes induced by an

infection, and occurs in all age groups [1]. Early onset sepsis (EOS) in newborns, defined as onset

of sepsis within 72 hours after birth, remains a clinical diagnostic and therapeutic challenge due

to its non-specific clinical presentation. This is associated with late discovery and undertreatment

of septic newborns or overtreatment with antibiotics of uninfected ones [2-4]. These diagnostic

and therapeutic problems arise because the pathophysiology of EOS is not completely

understood [3-5].

One of the pathological changes in septic patients is microvascular dysfunction leading to

increased vascular inflammation and leakage [6]. Vascular endothelial cells control these changes

through the Angiopoietin/Receptor Tyrosine Kinase (Tie)-2 endothelial receptor system, which

is severely disturbed in sepsis [6,7-9]. The system consists of the ligands Angiopoietin-1 (Ang-1)

and -2 (Ang-2) [9]. In health, Ang-1-Tie2 binding promotes intracellular Tie-2 phosphorylation,

which prevents the occurrence of vascular inflammation and vascular leakage [10]. During sepsis,

Ang-2 dose dependently competes with Ang-1, which inhibits Tie-2 phosphorylation and induces

destabilizing vascular inflammation and leakage [11,12]. In sepsis in children and adults, higher

Ang-2 levels and Ang-2/Ang-1 ratios in blood are associated with presence, severity and outcome of

sepsis, while changes in Ang-1 levels are less uniformly present [13-17]. To date, there is insufficient

knowledge if disturbances in the Angiopoietin/Tie2 system also reflect the activation state of

the endothelium during EOS in newborns [18]. Furthermore, no data exists on the Angiopoietins

during EOS from non-Western countries, such as Suriname.

Therefore, we studied the levels and behavior of Ang-1 and Ang-2 at start of antibiotic

treatment and at reevaluation between 48 to 72 hours in Surinamese newborns with suspected

EOS. We hypothesized that lower Ang-1 and higher Ang-2 and Ang-2/Ang-1 protein ratio were

associated with blood culture positive EOS.

MATERIALS AND METHODSStudy Design and Subjects

A prospective observational cohort study was performed at the neonatal care facility of

the Academic Pediatric Center Suriname at the Academic Hospital Paramaribo. Patients

were included in a 14-month period between April 1st 2015 and May 31st 2016. Newborns with

a gestational age (GA) equal to or above 34 weeks in whom antibiotics were started within the first

72 hours of life for suspected EOS were included. Excluded were neonates of whom no serum was

obtained or not enough information was available after the study period to confirm outcomes.

Written informed consent was obtained from at least one parent for the use of residual serum

and clinical information. The study protocol was approved by the Surinamese Medical-Ethical

Board (VG-021-14A) and was made available on clinicaltrials.gov (Trial registration: NCT02486783

registered 27/6/2015).

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

122

7

Clinical Protocol

For all newborns, the standard local protocol for the management of suspected EOS was followed.

This included the start of antibiotics after blood collection for culture and serial laboratory testing

of infectious parameters (t=0). Intravenous ampicillin (50-75 mg/kg/day) and gentamycin (5 mg/

kg/day) were started based on the presence of maternal risk factors for infection (i.e., positive

group B streptococcus culture, (premature) prolonged rupture of membranes, intrapartum

fever or intrapartum antibiotics) and/or clinical signs of infection of the newborn. Controls

were newborns without signs of infection receiving blood draws for hyperbilirubinemia. In these

controls, no antibiotics were started. Newborns in whom antibiotics were started were divided

in two groups based on blood culture result: 1) blood culture negative EOS and 2) blood culture

positive EOS.

Data Collection

For all newborns maternal information (i.e., history, pregnancy complications (i.e., presence of

diabetes mellitus, pregnancy-induced hypertension (PIH) or preeclampsia (PE)) and maternal risk

factors for infection) were recorded, along with gestational age (if unknown according to Ballard),

Apgar scores, birth weight, gender, ethnicity, results from laboratory testing (white blood cell

counts and CRP levels), duration of antibiotic treatment, blood culture results, hospital course,

and mortality.

Sample Collection, Preparation and Analysis

Blood samples were collected in serum microtainers using standard blood collection during

the insertion of a venous cannula. This time point was labeled t=0. After 48-72 hours of treatment

with antibiotics a second blood sample was obtained using capillary collection. This time point

was labeled t=48-72. CRP and hematological parameters were determined routinely at the clinical

laboratory of the Academic Hospital Paramaribo. Blood was allowed to clot at room temperature

and serum was separated by centrifugation at 2,300xg for 8 minutes, the serum was harvested

and residual sample was stored at -80°C until further analysis. Frozen samples were transported

on dry ice from Suriname to the Netherlands. For analysis, the samples were thawed on ice

and immediately analyzed. Measurement of levels of Ang-1 and Ang-2 was performed using

the Human Luminex Screening Assay LXSAH (R&D systems, Minneapolis, MN, USA) according

to the manufacturer’s instructions. We determined inter-assay coefficients of variation (CV) and

accepted a maximum of 20%. Median inter-assay CV ranged from 7.3% to 10.5% for Ang-1 and 4.6%

to 10.3% for Ang-2, respectively.

Statistical Analysis

Categorical variables were presented as numbers and percentages with 95% CI and compared

with chi-square. Continuous variables were presented as median and interquartile range (IQR)

Due to the nonparametric nature of the data a Mann-Whitney or Kruskal-Wallis test with Dunn’s

correction for multiple comparisons was used for analysis of continuous variables. Spearman’s

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

123

7

rho was used to assess bivariable associations between CRP levels and Ang-1 and Ang-2 levels,

respectively. P-values <0.05 were considered statistically significant. All analyses were performed

using Prism version 7.0a (Graphpad Software Inc., San Diego, CA, USA).

RESULTSDemographics

Of 101 eligible newborns 8 newborns were excluded for incomplete clinical information and

2 for insufficient serum. For the 91 included newborns demographics are given in Table 1. Birth

weight, age at presentation, Apgar score and clinical course at t=48-72h were distributed unevenly

amongst the three groups. Six (8.5%; 95% CI 3.9-17.2) newborns receiving antibiotic treatment had

a positive blood culture (all gram-negative bacteria, Klebsiella pneumoniae (n=2), Enterobacter

cloacae (n=2) and Escherichia coli (n=2). Newborns with EOS received respiratory and circulatory

support more often than controls (P<0.001). A total of five newborns with EOS died. White

blood cell, neutrophil and trombocyte counts and CRP levels were not different between groups

(Table 2).

Levels of Angiopoietins

At t=0, median levels of Ang-1 were significantly lower in blood culture positive EOS (28.3 (28.0)

ng/mL) versus controls (77.4 (65.2) ng/mL), P<0.01 (Table 2) (Figure 1A). Median Ang-2 levels

were higher in blood culture EOS (21.1 (13.3) ng/mL) versus controls (10.2 (1.9) ng/mL), P<0.001,

respectively) (Figure 1B). The Ang-2/Ang-1 protein ratio was higher in blood culture positive EOS

(median (IQR) 0.77 (0.77) versus controls (median (IQR) 0.13 (0.13) (P<0.01) (Figure 1C). There was

no difference in median levels of Ang-1, Ang-2 and Ang-2/Ang-1 protein ratio between blood

culture negative EOS and controls.

At t=48-72h, median Ang-1 levels had decreased 21-fold in blood culture positive EOS from

levels at t=0 (P=0.10), while median Ang-2 levels remained high (P=0.99) (Table 2) (Figure 1A-B).

Median levels of Ang-1, Ang-2 and Ang-2/Ang-1 protein ratio were not different when comparing

blood culture positive or blood culture negative EOS with controls.

Levels of Ang-1 and Ang-2 were tested for dependency on timing of first blood draw (t=0) after

birth. For controls and EOS (blood culture negative plus blood culture positive EOS) median levels

at t=0 were not different between newborns if t=0 was before 24 hours or between 24-72 hours

after birth (Figure 2A-B).

Because CRP levels at t=48-72h increased from levels at t=0 in blood culture negative

and positive EOS (Table 2), correlation of Ang-1 and Ang-2 with CRP was assessed amongst 44

newborns with blood culture negative (n=42) and positive (n=2) EOS in whom all data had been

recorded (Figure 3A-B). Lower median Ang-1 (rho -0.46; 95% CI -0.67 to -0.19), but not higher

Ang-2, correlated with higher CRP at t=48-72h.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

124

7

Table 1. Descriptive statistics of the study group (n=91)

Controls

(n=20)

Early Onset Sepsis

P-value

Blood Culture

Negative (n=65)

Blood Culture

Positive (n=6)

Pregnancy, n (%) Complications1

Chorioamnionitis2

3 (15)

0

16 (25)

18 (28)

1 (17)

0

0.63

Mode of delivery,

n (%)

Vaginal

Caesarean

12 (60)

8 (40)

46 (75)

19 (25)

4 (67)

2 (33)

0.54

Sex, n (%) Male

Female

9 (45)

11 (55)

29 (45)

36 (55)

5 (83)

1 (17)

0.19

Ethnicity, n (%) Maroon and Creole

Hindo-Surinamese

Other3

12 (60)

3 (15)

5 (25)

44 (68)

14 (21)

7 (11)

4 (67)

1 (17)

1 (17)

0.61

Gestational age,

n (%) (weeks)

34-37

37-40

≥40

1 (5)

14 (70)

5 (25)

22 (34)

30 (46)

13 (20)

0

4 (67)

2 (33)

0.06

Apgar score, n (%) <5 0 5 (8) 2 (33) 0.03

Birth weight, Median

(IQR) (grams)

3130 (700) 2840 (835) 3500 (906) 0.02

Age at presentation,

n (%) (hours)

<24

24-48

48-72

4 (20)

7 (35)

9 (45)

43 (66)

13 (20)

9 (14)

2 (33)

1 (17)

3 (50)

<0.01

Clinical course

(at 48-72h), n (%)

CPAP

Mechanical Ventilation

Cardiotonics

Mortality

0

0

0

0

9 (14)

7 (11)

5 (8)

3 (5)

0

2 (33)

1 (17)

2 (33)

<0.001

CPAP = continuous positive airway pressure; N/A = not applicable. 1 Presence of pregnancy-induced hypertension, preeclampsia or diabetes mellitus.2 Defined as intrapartum fever or administration of antibiotics.3 Includes: Javanese, Chinese, Caucasian and Amerindian.

DISCUSSIONIn this study, we investigated the serum levels of Ang-1 and Ang-2 to better understand the vascular

pathophysiology in near-term and term Surinamese newborns treated for EOS. Lower levels of

Ang-1, higher Ang-2, and a higher Ang-2/Ang-1 protein ratio in serum of newborns was associated

with blood culture positive EOS at start of antibiotic treatment. Levels of Ang-1 further decreased

over time in newborns with blood culture positive EOS and correlated negatively with higher levels

of CRP. These results indicate a role for the Angiopoietins in vascular inflammation during EOS in

Suriname. An estimated 5-10% of total EOS data is from non-Western countries such as Suriname,

while there is strong indication that over 90% of global deaths due to EOS occurs in these settings

[2,19,20]. Thus, our data add critical basic and clinical knowledge on the true global impact of EOS.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

125

7

Table 2. Infection biomarkers in baseline controls and newborns with suspected and blood culture positive

early onset sepsis

Time

Point n (%) Controls

Early Onset Sepsis

P-value

Blood Culture

Negative

Blood Culture

Positive

White blood cells (x109/L)1 t=0 88 (97) 15.3 (8.2) 17.5 (9.7) 21.9 (82.4) 0.68

Neutrophils (x109/L)1 t=0 72 (79) 7.1 (8.5) 9.2 (7.9) 10.2 (34.1) 0.57

Platelets (x109/L)1 t=0 83 (91) 235 (82) 239 (60) 74 (164.5) 0.07

C-reactive protein (mg/dL) t=0

t=48-72h

Delta

75 (82)

44 (48)

44 (48)

<0.5 (0)

N/A

N/A

<0.5 (0.7)

0.7 (1.8)

0.1 (1.3)

0.7 (4.8)

1.4 (16.3)

1.7 (3.4)

0.34

0.811

0.461

Angiopoietin-1 (ng/mL) t=0

t=48-72h

91 (100)

49 (54)

77.4 (65.2)

68.9 (44.5)

82.2 (45.7)

73.6 (67.3)

28.3 (28.0)

1.3 (16.1)

<0.01

0.021

Angiopoietin-2 (ng/mL) t=0

t=48-72h

91 (100)

49 (54)

10.2 (1.9)

9.9 (1.5)

11.2 (6.9)

11.8 (4.7)

21.1 (13.3)

19.0 (18.9)

<0.01

0.071

N/A = not applicable.

Data presented as median (IQR) and analyzed with a Kruskal-Wallis test between all groups or 1 with a Mann-Whitney test

between blood culture negative and positive EOS groups.

Our results of Ang-1 levels are in line with other studies that reported reduced Ang-1 levels

in children associated with septic shock and death [21,22]. The mechanism for low Ang-1 remains

poorly understood. While Ang-1 levels are low, the levels of its soluble ligand sTie-2 are higher

in the blood of septic patients. Soluble Tie 2 may acts as a decoy receptor binding Ang-1 with

high affinity, thereby decreasing its circulating levels. On the other hand, increasing Ang-1 levels,

thereby increasing endothelial Tie-2 receptor phosphorylation may help to inhibit vascular

inflammation and leakage. In a clinically relevant murine model, intravenous recombinant Ang-1

treatment was sufficient to improve sepsis-associated organ dysfunctions and survival time, most

likely by preserving endothelial barrier function [23].

Higher levels of Ang-2 may be reflective of vascular inflammation and vascular leakage.

Intravenous lipopolysaccharide injection in human volunteers, adult human sepsis, and secondary

infection in critically ill patients causes higher levels of circulating Ang-2 and higher Ang-2/Ang-1

ratios [24-28]. As intracellular Tie-2 phosphorylation cannot be assessed in patients, an increased

Ang-2/Ang-1 ratio might be predictive for reduced endothelial Tie-2 receptor phosphorylation

with subsequent vascular inflammation and leakage.

EOS can occur following colonization of the newborn with bacterial pathogens following

intra-uterine infection or in the birth canal during labor [4]. Two studies found higher maternal

and amniotic fluid levels of Ang-2 in cases of intra-uterine infection in at term and preterm birth

[29,30]. To our knowledge, placental Ang-2 crossing has not been described. The presence of

intra-uterine infection may result in EOS and cause subsequent suppression of neonatal levels

of Ang-1 and release of neonatal Ang-2 from endothelial cells. Our finding that levels of Ang-1

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

126

7

Ang

-2 (n

g/m

L)

Controls

A

B

C

Ang

-1 (n

g/m

L)A

ng-2

/Ang

-1

Blood Culture Positive EOS

t=0 (n=20)

t=48-72 (n=3)

*

Pt=0 <0.01Pt=48-72 =0.02

Pt=0 <0.01Pt=48-72 =0.07

Pt=0 <0.001Pt=48-72 <0.01

0

50

100

150

0

10

20

30

40

0.0

0.5

1.0

1.510

15

20

25

t=0 (n=65)

t=48-72 (n=43)

t=0 (n=6)

t=48-72 (n=3)

Blood Culture Negative EOS

*

*

Figure 1. Serum levels of Angiopoietin-1 and Angiopoietin-2 of controls and newborns with blood culture

negative and positive early onset sepsis (EOS). A: Angiopoietin-1 (Ang-1); B: Angiopoietin-2 (Ang-2); C:

Ang-2/Ang-1protein ratio; Data represent levels in serum sampled at t=0 (white bars) and t=48-72h (grey

bars) and are analyzed with a Kruskal-Wallis test with Dunn’s correction for multiple comparisons between

all groups at t=0 (Pt=0

) and at t=48-72 (Pt=48-72

). * P<0.05 when groups are separately compared to controls. Bars

represent median values and error bars interquartile range.

and Ang-2 are similar between infected newborns included directly after birth and after 24 hours

supports this hypothesis.

A remarkable finding in our study was that levels of Ang-1 in newborns were up to a 10-fold

higher, specifically in healthy newborns and those with blood culture negative EOS, than in

children or adults in earlier studies [13-17,21,22]. Placental levels of Ang-1 and Ang-2 are high

during pregnancy and then quickly drop after birth [31,32]. Only one earlier study compared both

antepartum and post caesarean maternal samples with neonatal umbilical cord blood samples

[32]. Ang-1 concentrations were significantly higher in umbilical samples, suggesting separate

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

127

7 <24h (n=4)

Ang

-2 (n

g/m

L)

A

B

Ang

-1 (n

g/m

L)

Controls

<24h (n=45)

24-72h (n=16)

24-72h (n=26)

EOS

Age at inclusion

0

50

100

150

0

5

10

15

20

Figure 2. Serum levels of Angiopoietin-1 and Angiopoietin-2 at inclusion in newborns included before

and after the first 24 hours of life. A: Angiopoietin-1 (Ang-1); B: Angiopoietin-2 (Ang-2); Data represent

pooled levels at t=0 from newborns considered uninfected (controls) and from newborns considered

infected (blood culture negative and positive EOS), included before 24h (light grey bars) versus 24-72h

(checked light grey bars) after birth. Data was analyzed with a Mann-Whitney test. Bars represent median

values and error bars interquartile range.

0 5 10 15 20 250

50

100

150

200

250

Ang

-1 (n

g/m

L)

0 5 10 15 20 250

10

20

30

40

CRP (mg/dL)

Ang

-2 (n

g/m

L)

A

B

rho -0.46; P<0.01

Figure 3. Correlations of serum levels of Angiopoietin-1 and Angiopoietin-2 with serum levels of

C-reactive protein in newborns with blood culture negative or positive early onset sepsis (EOS).

Correlations of CRP with A: Angiopoietin-1 (Ang-1); B: Angiopoietin-2 (Ang-2); Data represent levels of Ang-1,

Ang-2 and CRP in serum sampled at t=48-72h from newborns (in whom data on levels was universally available)

with blood culture negative (n=42) and positive (n=2) EOS. Spearman’s rho was used to assess correlations.

Correlation (rho) is given when significant.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

128

7

Angiopoietin regulation in the newborn. Animal models of pregnancy may help elucidate the exact

dynamics of Angiopoietins in newborns [33]. These animal models may also be instrumental in

detecting endothelial Tie-2 receptor phosphorylation in different microvascular beds.

From a clinical perspective, our findings indicate that serial measurement of Angiopoietins

may predict or exclude bacteremia in newborns before blood culture results are known. High

serial Ang-1 and low serial Ang2/Ang-1 ratio may be extra arguments to discontinue antibiotics,

alongside serial measurement of CRP. A known limitation to CRP is its slow synthesis limiting its

utility in early prediction of EOS. To overcome this issue, inflammatory mediators that precede

CRP synthesis, such as Interleukin (IL)-1ß, IL-6, IL-8 and Tumor-necrosis Factor (TNF)-a have been

of interest in EOS research [34-36]. These mediators have short half-lives, which limits their clinical

use and establishment of appropriate cut-off values. In our study, levels of Ang-1 remained high

in healthy and low in the sickest newborns at reevaluation 48-72 hours after start of antibiotics,

indicating persistent association with severity of disease over time and clinical utility. Additionally,

TNF-a has been shown to correlate with Ang-2 levels in adult patients with sepsis [24,37]. For these

reasons it would be interesting to evaluate temporal relations of the Angiopoietins with a panel of

inflammatory mediators, such as TNF-α, Il-6 and IL-1ß in EOS.

Our study has several limitations. First, our sample size was relatively small to assess relevance

of the Angiopoietins as clinical biomarkers and results may have been confounded by birth

weight and asphyxia, which were distributed unevenly amongst the groups. Second, as levels of

the Angiopoietins were determined with a Luminex Screening Assay we were unable to compare

levels with results from other studies measured with ELISA [21,28], and small sample volumes

acquired in neonates precluded assessment of other inflammatory mediators. Future studies

will focus on eliminating these limitations to enable us to validate the current observations in

newborns in Surinamese newborns.

In summary, our data show changes in the ligands of the Angiopoietin/Tie2 endothelial receptor

system Ang-1 and Ang-2 in EOS and support the hypothesis that increased vascular inflammation

and increased vascular leakage leads to microvascular dysfunction in the pathophysiology of EOS.

The potential impact of intra-uterine-infection deserves attention in future investigations to

further elucidate dynamics of Angiopoietins in newborns with and without EOS.

Abbreviations

EOS = early onset sepsis

Ang-1 = Angiopoietin-1

Ang-2 = Angiopoietin-2

Tie-2 = TEK tyrosine kinase endothelial-2 receptor

Conflict of Interest

The authors declare that they have no conflict of interest.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

129

7

Acknowledgments

RZ was supported by a stipend from the Thrasher Research Fund (TRF13064) and from Tergooi

Hospitals, Blaricum, The Netherlands. The authors acknowled the efforts of all employees of

the Clinical Laboratory of the Academic Hospital Paramaribo and the Central Laboratory of

Suriname, Paramaribo, Suriname, for assistance with sample storage, handling and transport.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

130

7

REFERENCES

1. Singer M, Deutschman CS, Seymour CW,

Shankar-Hari M, Annane D, Bauer M, Bellomo

R, Bernard GR, Chiche JD, Coopersmith CM,

Hotchkiss RS, Levy MM, Marshall JC, Martin

GS, Opal SM, Rubenfeld GD, van der Poll T,

Vincent JL, Angus DC. The Third International

Consensus Definitions for Sepsis and Septic

Shock (Sepsis-3). JAMA 2016, 315(8):801-10.

2. Schrag SJ, Farley MM, Petit S, Reingold A,

Weston EJ, Pondo T, Hudson Jain J, Lynfield.

Epidemiology of Invasive Early-Onset Neonatal

Sepsis, 2005 to 2014. Pediatrics 2016, 138(6).

3. van Herk W, Stocker M, van Rossum AM.

Recognising early onset neonatal sepsis: an

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77-82.

4. Simonson KA, Anderson-Berry AL, Delair SF,

Davies HD. Early-onset neonatal sepsis. Clin

Microbiol Rev 2014, 27(1):21-47.

5. Hofer N, Zacharias E, Müller W, Resch B. An

update on the use of C-reactive protein in

early-onset neonatal sepsis: Current insights

and new tasks. Neonatology 2012, 102:25–36.

6. Aird WC. The role of the endothelium in

severe sepsis and multiple organ dysfunction

syndrome. Blood 2003, 101(10):3765-77.

7. van Meurs M, Kümpers P, Ligtenberg JJ,

Meertens JH, Molema G, Zijlstra JG. Bench-to-

bedside review: Angiopoietin signalling in critical

illness - a future target? Crit Care 2009, 13(2):207.

8. Parikh SM. Dysregulation of

the angiopoietin-Tie-2 axis in sepsis and ARDS.

Virulence 2013, 4(6):517-24.

9. Kurniati NF, Jongman RM, vom Hagen F,

Spokes KC, Moser J, Regan ER, Krenning

G, Moonen JR, Harmsen MC, Struys MM,

Hammes HP, Zijlstra JG, Aird WC, Heeringa P,

Molema G, van Meurs M. The flow dependency

of Tie2 expression in endotoxemia. Intensive

Care Med 2013, 39(7):1262-71.

10. Kim M, Allen B, Korhonen EA. Opposing actions

of angiopoietin-2 on Tie-2 signaling and FOXO1

activation. J Clin Invest 2016, 126(9):3511-25.

11. Yuan HT, Khankin EV, Karumanchi SA, Parikh

SM. Angiopoietin 2 is a partial agonist/

antagonist of Tie2 signaling in the endothelium.

Mol Cell Biol 2009, 29(8):2011-22.

12. Van der Heijden M, van Nieuw Amerongen

GP, van Hinsbergh VW, Groeneveld AB.

The interaction of soluble Tie2 with

angiopoietins and pulmonary vascular

permeability in septic and nonseptic critically

ill patients. Shock 2010, 33(3):263-8.

13. Giuliano JS Jr, Tran K, Li FY, Shabanova V,

Tala JA, Bhandari V. The temporal kinetics of

circulating angiopoietin levels in children with

sepsis. Pediatr Crit Care Med 2014, 15(1):e1-8.

14. Wang K, Bhandari V, Giuliano JS Jr.

Angiopoietin-1, angiopoietin-2 and bicarbonate

as diagnostic biomarkers in children with severe

sepsis. PLoS One 2014, 9(9):e108461.

15. Lymperopoulou K, Velissaris D, Kotsaki A,

Antypa E, Georgiadou S, Tsaganos T, Koulenti

D, Paggalou E, Damoraki G, Karagiannidis N,

Orfanos SE. Angiopoietin-2 associations with

the underlying infection and sepsis severity.

Cytokine 2015, 73(1):163-8.

16. Ricciuto DR, dos Santos CC, Hawkes M, Toltl

LJ, Conroy AL, Rajwans N, Lafferty EI, Cook DJ,

Fox-Robichaud A, Kahnamoui K, Kain KC, Liaw

PC, Liles WC. Angiopoietin-1 and angiopoietin-2

as clinically informative prognostic biomarkers

of morbidity and mortality in severe sepsis. Crit

Care Med 2011, 39(4):702-10.

17. Fang Y, Li C, Shao R, Yu H, Zhang Q, Zhao L.

Prognostic significance of the angiopoietin-2/

angiopoietin-1 and angiopoietin-1/Tie-2 ratios

for early sepsis in an emergency department.

Crit Care 2015, 14(19):367.

18. Mussap M, Cibecchini F, Noto A, Fanos

V. In search of biomarkers for diagnosing

and managing neonatal sepsis: the role

of angiopoietins. J Matern Fetal Neonatal

Med 2013, 26(2):24-6.

19. Edmond KM, Kortsalioudaki C, Scott S, Schrag

SJ, Zaidi AK, Cousens S, Heath PT. Group B

streptococcal disease in infants aged younger

than 3 months: systematic review and meta-

analysis. Lancet 2013, 379(9815):547-56.

20. Lawn JE, Cousens S, Zupan J; Lancet

Neonatal Survival Steering Team. 4 million

neonatal deaths: when? Where? Why?

Lancet 2005, 365(9462):891-900.

AN

GIO

POIETIN

S IN SU

RINA

MESE N

EWBO

RNS

131

7

21. Mankhambo LA, Banda DL; IPD Study Group,

Jeffers G, White SA, Balmer P, Nkhoma S,

Phiri H, Molyneux EM, Hart CA, Molyneux

ME, Heyderman RS, Carrol ED. The role of

angiogenic factors in predicting clinical

outcome in severe bacterial infection in

Malawian children. Crit Care 2010, 14(3):R91.

22. Guiliano JS, Lahni PM, Harmon K, Wong

HR, Doughty LA, Carcillo JA, Zingarelli

B, Sukhatme VP, Parikh SM, Wheeler DS.

Admission angiopoeitin levels in children with

septic shock. Shock 2007, 28(6):650-654.

23. David S, Park JK, Meurs Mv, Zijlstra JG, Koenecke

C, Schrimpf C, Shushakova N, Gueler F, Haller H,

Kümpers P. Acute administration of recombinant

Angiopoietin-1 ameliorates multiple-organ

dysfunction syndrome and improves survival in

murine sepsis. Cytokine 2011, 55(2):251-9.

24. Fiedler U, Reiss Y, Scharpfenecker M, Grunow

V, Koidl S, Thurston G, Gale NW, Witzenrath

M, Rosseau S, Suttorp N, Sobke A, Herrmann

M, Preissner KT, Vajkoczy P, Augustin HG.

Angiopoietin-2 sensitizes endothelial cells to

TNF-alpha and has a crucial role in the induction

of inflammation. Nat Med 2006, 12(2):235-9.

25. Parikh SM, Mammoto T, Schultz A, Yuan

HT, Christiani D, Karumanchi SA, Sukhatme

VP. Excess circulating angiopoietin-2 may

contribute to pulmonary vascular leak in sepsis

in humans. PLoS Med 2006, 3(3):e46.

26. Kümpers P, van Meurs M, David S, Molema G,

Bijzet J, Lukasz A, Biertz F, Haller H, Zijlstra JG.

Time course of angiopoietin-2 release during

experimental human endotoxemia and sepsis.

Crit Care 2009, 13(3):R64.

27. David S, Mukherjee A, Ghosh CC, Yano M,

Khankin EV, Wenger JB, Karumanchi SA, Shapiro

NI, Parikh SM. Angiopoietin-2 may contribute

to multiple organ dysfunction and death in

sepsis. Crit Care Med 2012, 40(11):3034-41.

28. Van Vught LA, Wiewel MA, Hoogendijk AJ,

Frencken JF, Scicluna BP, Klein Klouwenberg

PM, Zwinderman AH, Lutter R, Horn J, Schultz

MJ, Bonten MMJ, Cremer OL, van der Poll

T. The Host Response in Sepsis Patients

Developing Intensive Care Unit-acquired

Secondary Infections. Am J Respir Crit Care

Med 2017, 196(4):458-470.

29. Pacora P, Romero R, Chaiworapongsa T,

Kusanovic JP, Erez O, Vaisbuch E, Mazaki-Tovi

S, Gotsch F, Jai Kim C, Than NG, Yeo L, Mittal

P, Hassan SS. Amniotic fluid angiopoietin-2

in term and preterm parturition, and intra-

amniotic infection/inflammation. J Perinat

Med 2009, 37(5):503-11.

30. Buhimschi CS, Bhandari V, Dulay AT, Thung

S, Razeq SS, Rosenberg V, Han CS, Ali UA,

Zambrano E, Zhao G, Funai EF, Buhimschi IA.

Amniotic fluid angiopoietin-1, angiopoietin-2,

and soluble receptor tunica interna endothelial

cell kinase-2 levels and regulation in normal

pregnancy and intraamniotic inflammation-

induced preterm birth. J Clin Endocrinol

Metab 2010, 95(7):3428-36.

31. Kappou D, Sifakis S, Konstantinidou A,

Papantoniou N, Spandidos DA. Role of

the angiopoietin/Tie system in pregnancy. Exp

Ther Med 2015, 9(4):1091-1096.

32. Keikkala E, Hytinantti T, Wathén KA, Andersson

S, Vuorela P. Significant decrease in maternal

serum concentrations of angiopoietin-1

and -2 after delivery. Acta Obstet Gynecol

Scand 91(8):917-22, 2012.

33. Reynolds LP, Borowicz PP, Vonnahme KA,

Johnson ML, Grazul-Bilska AT, Wallace JM, Caton

JS, Redmer DA. Animal models of placental

angiogenesis. Placenta 2005, 26(10):689-708.

34. Machado JR, Soave DF, da Silva MV, de

Menezes LB, Etchebehere RM, Monteiro ML,

dos Reis MA, Corrêa RR, Celes MR. Neonatal

sepsis and inflammatory mediators. Mediators

Inflamm 2014, 269681..

35. Khaertynov KS, Boichuk SV, Khaiboullina SF,

Anokhin VA, Andreeva AA, Lombardi VC,

Satrutdinov MA, Agafonova EA, Rizvanov AA.

Comparative Assessment of Cytokine Pattern

in Early and Late Onset of Neonatal Sepsis. J

Immunol Res 2017, 8601063.

36. Fattah MA, Omer AF, Asaif S, Manlulu R, Karar T,

Ahmed A, Aljada A, Saleh AM, Qureshi S, Nasr A.

Utility of cytokine, adhesion molecule and acute

phase proteins in early diagnosis of neonatal

sepsis. J Nat Sci Biol Med 2017, 8(1):32-39.

37. Benest AV, Kruse K, Savant S, Thomas M,

Laib AM, Loos EK, Fiedler U, Augustin HG.

Angiopoietin-2 is critical for cytokine-induced

vascular leakage. PLoS One 2013, 8(8):e70459.

8 Analyzing Neutrophil Morphology, Mechanics, and Motility in Sepsis: Options and Challenges for Novel

Bedside Technologies

Rens Zonneveld, Grietje Molema, Frans B. Plötz

Critical Care Medicine 2016, 44:218-28

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

134

8

ABSTRACTObjective

Alterations in neutrophil morphology (size, shape and composition), mechanics (deformability),

and motility (chemotaxis and migration) have been observed during sepsis. We combine

summarizing features of neutrophil morphology, mechanics, and motility that change during

sepsis with an investigation into their clinical utility as markers for sepsis through measurement

with novel technologies.

Data Sources

We performed an initial literature search in MEDLINE using search-terms ‘neutrophil’, ‘morphology’,

‘mechanics’, ‘dynamics’, ‘motility’, ‘mobility’, ‘spreading’, ‘polarization’, ‘migration’, ‘chemotaxis’.

We then combined the results with ‘sepsis’ and ‘septic shock’. We scanned bibliographies of

included articles to identify additional articles.

Study Selection and Data Extraction

Final selection was done after the authors reviewed recovered articles. We included articles based

on their relevance for our review topic.

Data Synthesis

When compared to resting conditions, sepsis causes an increase in circulating numbers of larger,

more rigid neutrophils that show diminished granularity, migration and chemotaxis. Combined

measurement of these parameters could provide a more complete view on neutrophil phenotype

manifestation. For that purpose, sophisticated automated hematology analysers, microscopy and

bedside microfluidic devices provide clinically feasible, high throughput, and cost limiting means.

Conclusions

We propose that integration of features of neutrophil morphology, mechanics and motility with

these new analytical methods can be useful as markers for diagnosis, prognosis and monitoring of

sepsis and may even contribute to basic understanding of its pathophysiology.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

135

8

INTRODUCTIONNeutrophils play an essential role during infection and sepsis. While they actively remove invading

pathogens during infection, they contribute to the development of septic shock and multi-organ

failure through their excessive inflammatory activation, aberrant recruitment, and dysregulated

interactions with the vascular endothelium [1-3]. Alterations in neutrophil morphology (size, shape

and composition), mechanics (deformability), and motility (chemotaxis and migration) have been

observed during sepsis in experimental and clinical studies. For example, during sepsis neutrophils

become larger and less granular, which is associated with worse outcome [4-6]. Additionally,

neutrophils from septic patients are more resistant to mechanical forces [7] and show slower and

diminished migration and chemotaxis when compared to neutrophils from healthy controls [8,9].

To date, the exact roles of such alterations in neutrophils during sepsis remain unclear.

Novel technologies that facilitate detailed high-throughput measurement of these alterations

are currently available. Combinatory use of such methods may help to integrate features of

morphology, mechanics and motility into a model of specific neutrophil phenotypes. For example,

during infection and sepsis, these phenotypes may correlate with clinical features, and predict

outcomes. Based on the extent of change of these features, we may be able to distinguish a septic

phenotype from a phenotype that corresponds with mere local infection. Ideally, the neutrophils

then return to a baseline phenotype after successful treatment or recovery. Analysis of the above

mentioned phenotypes could furthermore improve our understanding of neutrophil physiology

and sepsis pathogenesis.

Therefore, we combined summarizing the current literature on features of neutrophil

morphology, mechanics and motility that change during sepsis with an investigation into their

clinical utility as markers for sepsis through measurement with novel technologies.

POTENTIAL FEATURES OF NEUTROPHIL PHENOTYPESIn this section we focus on experimental and clinical evidence for alterations in morphology,

mechanics and motility of neutrophils during sepsis and correlations with severity of disease and

mortality. Table 1 summarizes these properties, along with established and novel methodologies

that can be used to measure them.

Morphology

Morphological features of neutrophils comprise size, shape and (intracellular) composition.

The bone marrow contains and retains neutrophil precursor cells representing different stages

of maturation (myeloblasts, promyelocyte, myelocyte, and metamyelocyte, respectively), and

neutrophils with specific, morphologically distinguishable nuclear compositions, such as immature

‘band’ neutrophils, and mature polymorphonuclear ‘segmented’ neutrophils [3,10]. During sepsis,

these cell types are all released into the circulation. An increased ratio of immature to mature

neutrophils in the blood correlates with the presence of, and discriminates between, infection

and sepsis [11,12]. Microscopic examination of traditional peripheral blood smears reveals that

circulating neutrophil precursors and immature neutrophils are larger than senescent neutrophils.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

136

8

Larger neutrophils may also result from inflammatory activation of circulating neutrophils in

the blood stream. For example, Hoffstein et al. found larger cell sizes after short in vitro incubation

with bacterial derived Formyl-Methionyl-Leucyl-Phenylalanin (fMLP) [13], possibly reflecting

neutrophil responses that also happen in vivo. This increase in cell size was due to fusion of granule

membranes with the cell membrane after degranulation. The clinical relevance of increases in size,

shape, volume and granularity, of neutrophils during sepsis is further revealed in studies using

devices such as automated hematology analyzers or flow cytometers, which are discussed below.

Blood smears also reveal neutrophil changes in intracellular composition after inflammatory

stimulation such as the occurrence of cytoplasmic vacuoles, Döhle bodies and toxic granules.

Furthermore, both experimental and clinical studies suggest that based on morphological features

of apoptosis (e.g., nuclear condensation, perinuclear chromatin aggregation and membrane

blebbing), the degree of neutrophil apoptosis is inversely related to sepsis severity [14-16].

Additionally, a range of experimental studies and some clinical data show association of increased

formation of Neutrophil Extracellular Traps (NETs) with sepsis [10,16-19]. NETs are thought to be

a defense mechanism against invading pathogens and present extracellular presence of DNA,

covered with histones, granular proteins and enzymes. Much of the evidence on NETs is based

on ex vivo stimulation of neutrophils with cytokines (Tumor Necrosis Factor (TNF-β), IL-8 or IL1β),

plasma from SIRS or septic patients, bacteria, or artificial stimulation with Phorbol 12-myristate

13-acetate (PMA) to form NETs [10,17]. Staining blood smears with immunofluorescently labeled

antibodies (e.g., against neutrophil elastase or DNA) can identify NETs in patients [18,19]. At

present, this analysis is not clinically feasible yet and data reported so far was not sufficient to

discriminate sepsis from other causes of critical illness [18,19].

Table 1. Classic and novel methods for the assessment of neutrophil morphological, mechanical and dynamic

changes in sepsis

Cell Property

Measured Classic Methods Novel Methods Clinical Implementation

Morphology Size, Volume,

Granularity

Subcellular

structures

Immature count

Blood smears

EM

Automated hematology

analysers

Standardized flow

cytometry

Microscopy

Automated hematology

analysers

Standardized flow

cytometry

Apoptosis

NETosis

IF Microscopy

EM

None IF measurement of NETs in

blood smears

Mechanics Deformability Sequestration

assays

Micropipette

assays

Magnetic twisting

cytometry

Atomic Force Microscopy

None yet

Motility Migration

Chemotaxis

Transwell assays Time Lapse Microscopy

Microfluidic

Devices, K.O.A.L.A.

Bedside Microfluidic

Devices / K.O.A.L.A.

IF = immunofluorescence; EM = Electron Microscopy; K.O.A.L.A. = Kit-on-a-Lid-Assay

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

137

8

Mechanics

In vivo, in order to pass through the microvasculature (diameter 5-6 μm), neutrophils (diameter

7-9 μm) have to be deformable (i.e., flexible or less rigid/stiff). Deformability of neutrophils was

investigated in relation to their pulmonary microvascular sequestration in experimental mouse

models of sepsis and acute respiratory distress syndrome (ARDS), and in vitro in micropore transit

assays [20-25]. Collectively, these studies concluded that a decrease in deformability of activated

neutrophils prevented them from traversing through the microvasculature or micropores, or

prolonged their transit time in in vitro assays. Other determinants that alter with inflammatory

activation and that contribute to increased transit time, such as adhesion (e.g., to endothelium in

vivo or to substrates coated with immobilized adhesion molecules in laboratory setting) were often

not taken into account. Only one in vivo study in rats described sequestration due to increased

stiffness of neutrophils, which still occurred after pharmacological blocking of neutrophil integrins

that are necessary for proper adhesion of neutrophils to the endothelium, indicating stiffness as

the main determinant [25].

When deformability is measured ex vivo, some differences exist between different subtypes of

neutrophils. First, immature neutrophils in the bone marrow are reported to be less deformable

compared to circulating neutrophils [26]. Second, inflammatory stimulation of circulating

neutrophils causes decreased deformability of these cells [27]. This implies that an overall decreased

deformability of neutrophils in sepsis may be the resultant of increased rigidity of different subsets

(i.e., immature or mature, quiescent or activated). In one clinical study neutrophils from septic

patients showed substantially less deformability when compared to neutrophils from healthy

volunteers [7]. Interestingly, neutrophils from patients with septic shock and ARDS were even less

deformable than neutrophils from patients that had sepsis [7]. Neutrophils from septic neonates

were shown to be substantially more rigid than those from healthy neonates [28]. Since also

the percentages of circulating immature neutrophils in septic neonates were increased, the data

implied that both immature and activated neutrophils were important contributors to overall

stiffness of the whole population.

Motility

Upon inflammatory activation in vivo, neutrophils adhere to activated vascular endothelial cells

before migrating on and through the endothelium into the underlying tissues [3,10]. These

responses are referred to as adhesion, migration and chemotaxis, respectively [3,10]. Translational

studies have revealed that neutrophils from septic patients show supranormal adhesion to in vitro

cultured endothelium, yet reduced migration [3,30-36]. Migration and chemotaxis of neutrophils

have been quantitatively studied in ex vivo experimental settings in which patients’ blood

derived neutrophils were allowed to migrate across micropore membranes in transwells towards

a gradient of a chemoattractant (e.g., fMLP, casein or IL-8) for a defined length of time, followed

by fixation and microscopic quantification [30-36]. Two early studies addressed the potential

relevance of impaired migration of neutrophils in post-surgical patients and found a significantly

lower total number of migrated neutrophils from patients who developed sepsis [33,34]. Results

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

138

8

from neutrophils from controls treated with serum or plasma from septic patients were similar in

these studies. Two decades later these observations were confirmed in a cohort of septic patients,

which revealed that the degree of impairment was associated with non-survival [35]. Furthermore,

neutrophils from breast cancer patients who developed bacterial infection after chemotherapy

showed less migration when compared to cells from non-infected breast cancer patients

[36]. Finally, Najma et al. found a 72% reduction in neutrophil migration into in situ secondary

inflammatory sites in septic patients [37].

Although these studies were informative about the level of impairment of migration of

neutrophils from septic patients, detailed dynamics underlying these observations could at

that time not be obtained. For this purpose, advances in intra-vital and live cell microscopy and

microfluidics now provide tools for a qualitative description of dynamic events [38-41]. These

methods can give direct visual evidence of deviations from normal behavior during these events,

specifically with regard to how septic neutrophils shape (including polarization and spreading)

and move (including direction and speed) in comparison to healthy neutrophils, as will be

discussed below.

Summary

Figure 1 shows a schematic representation of in vivo neutrophils under resting conditions, infection

and sepsis. Sepsis causes an increase in their circulating numbers (including increased numbers of

immature neutrophils and precursors), size and stiffness (either as a direct effect of activation or

because of the increase in percentage of larger and stiffer immature neutrophils), and a decrease

in migration/chemotactic responses. Septic neutrophils are also prone to produce NETs and show

less apoptosis, each to be discerned based on their own specific morphological features.

Whilst most studies focus on one particular feature that changes, some indicate that they are

dependent on, or the result of, each other, and, as such, part of an integrated immune response.

For example, 1) not only decreased neutrophil deformability, but also larger cell size and adhesion

to the endothelium caused microvascular sequestration [28]; 2) the ability of neutrophils to

migrate was inversely related to increased adhesion and spreading (and concomitant actin re-

arrangements) [41]; 3) larger cell sizes were the result of degranulation and membrane addition,

which was associated with inflammatory activation [13]; 4) while immature neutrophils were larger

and less granular, they were more resistant to apoptosis and prone to the formation of NETs [17].

Furthermore, an integrated series of changes in neutrophils was proposed in studies describing

transcriptional profiles of neutrophils and other leukocytes from septic patients and during

endotoxemia [42,43]. Data from these studies featured neutrophils that showed simultaneous

upregulation of genes involved in actin cytoskeleton signaling, migration and extravasation, and

downregulation of genes participating in apoptosis and antigen presentation. The signature of

this ‘genomic storm’ may underlie the changes observed in neutrophil morphology, mechanics

and motility during sepsis, which emphasizes the likelihood that measurement of the latter can

provide a more complete view on neutrophil phenotype manifestation. In the next section we will

discuss novel methodologies and techniques that facilitate the measurement of these changes for

clinical use in the near future.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

139

8

NOVEL METHODS FOR IDENTIFICATION OF NEUTROPHIL PHENOTYPES This section describes current methodology and advances in the development of new methods

to include features of neutrophil morphology, mechanics and motility in the clinical assessment of

sepsis (summarized in Table 1).

Automated Measurement of Neutrophil Morphology

As discussed above, distinct neutrophil morphological features associated with inflammation

or sepsis have been well defined for over 100 years in microscopic analysis of peripheral blood

smears. Reproducibility and prognostic value of this test for sepsis, however, remains poor because

of the manual analysis that requires experienced technicians and is subject to human bias [44].

Additionally, analysis of neutrophils in blood smears does not allow for discrimination between

local infection and systemic inflammation (e.g., associated with sepsis), nor between bacterial,

viral or sterile inflammation. High-throughput automated hematology analyzers (AHA) and

standardized clinical flow cytometry may provide means to determine a number of morphological

parameters of many neutrophils at the same time and thus may minimize these limitations.

Automated hematology analyzers

Automated hematology analyzers can measure all cell types in peripheral blood of patients.

Based on electrical properties (magnetics, impedance) and scatter and absorption of light, they

provide detailed information on leukocyte subpopulations and the collectively named Volume,

Conductivity and Scatter (VCS) parameters [45,46]. VCS parameters can be measured in whole

blood without the need for isolation of neutrophils, extensive sample handling and consequent

risk of inducing activation of neutrophils. Some disadvantages of AHA include false identification

of clumps of cells or platelets as large (e.g., immature) neutrophils. The latest generation of

AHA minimizes these limitations, measures more parameters, and can even be equipped with

lasers for measurement of cell surface markers detected by for example immunofluorescently

labeled antibodies.

The performance of AHA analyzers for the diagnosis of sepsis was tested in a range of clinical

studies, which specifically focused on VCS parameters [4-6,45-48], immature neutrophil counts

[12,49-53], and, more recently, the delta neutrophil index (DNI) (i.e., a reflection of the immature

neutrophil population established by measuring the neutrophil differentials for MPO-activity and

nuclear lobularity [54-59]). This work showed that septic patients usually present with significantly

different VCS parameters (i.c., larger volumes and lower scatter, indicating degranulation), and

a larger fraction of immature neutrophils, also reflected by a higher DNI (Table 2). Important

findings in these studies were that VCS parameters and DNI could serve as early indicators of

sepsis (including neonatal sepsis) [49,51,52-55,58,59], that more aberrant values were associated

with severity of disease and mortality [46-48,52,56], and that upon treatment or recovery these

values returned to baseline [58,59].

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

140

8

A. Resting

B. Local infection

C. Sepsis

InflamedTissue

BoneMarrow

BoneMarrow

BoneMarrow

Interstitium

i: Hyperadhesion with potential for EC damage & Vascular Leak

ii: Sequestrationwithout

EC damage

Figure 1. Schematic representation of

neutrophil behavior in vivo. Under resting

conditions A: neutrophils are retained

in the bone marrow and circulation, and

only have short-term, reversible tethering

and rolling interactions with the vascular

endothelium. During infection/inflammation

B: the circulating inflammatory mediators

stimulate release of neutrophils and

precursors from the bone marrow into

the circulation. Upon encountering activated

endothelial cells, neutrophils upregulate/

activate adhesion molecules (including

integrins) on their cell surface. Neutrophils

then engage into coordinated interactions

with the endothelium (i.e., the leukocyte

adhesion cascade; see reference 29 for

detailed description of these events) and

ultimately transmigrate into the underlying

inflamed tissue. As part of a hypothesis on

the role of neutrophils under septic conditons

C: neutrophils can become activated by

high levels of circulating bacteria derived

agonists, such as endotoxin, and cytokines

such as Tumor Necrosis Factor (TNF- α) (also

known as the cytokine storm). Earlier, albeit

limited, data suggest that such activation

could i: promote excessive upregulation

of integrins on neutrophil cell surfaces,

driving hyperadhesion to the endothelium

with potential for collateral endothelial

damage (Reviewed in reference 3).

However, no direct evidence of structural

damage to the endothelium after neutrophil

adhesion during sepsis is currently available.

Furthermore, ii: re-release of neutrophils

into the circulation after periods of

sequestration/margination (e.g., due to

decreased deformability or size discrepancy

between relatively large diameter of

neutrophils versus the small diameter of

capillaries) without structural damage to

the endothelium or other tissues has also

been described (Reviewed in reference 82).

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

141

8

Tab

le 2

. Clin

ical

stu

dies

usi

ng a

uto

mat

ed h

emat

olo

gy

anal

ysis

of n

eutr

oph

il m

orp

holo

gy

in s

epsi

s

Art

icle

[re

f.]

Pop

ulat

ion

Para

met

ers

Co

ntr

ol1

Sep

tic1

P-va

lue

Co

rrel

atio

nA

nal

yzer

Cel

ik e

t al

. 20

12 [4

]N

eona

tes

Vo

lum

e

Co

nduc

tivi

ty

Scat

ter

148.

4 (1

1.1)

161.

5 (9

.1)

129

(10

.3)

170

.2 (

18.9

)

155.

9 (1

1.9)

125.

5 (1

1.8

<0.0

5

<0.0

5

<0.0

5

Earl

y d

iagn

osi

s o

f sep

sis

Co

ulte

r

LH 7

80

Mar

di e

t al

. 20

10 [5

]A

dul

tsV

olu

me

Co

nduc

tivi

ty

Scat

ter

139

(6.6

)

143

(3.3

)

144

(12.

1)

159

(16.

2)

146

(4.3

)

137

(12.

0)

<0.0

01

0.0

13

<0.0

01

Dia

gno

sis

of s

epsi

s

Dis

crim

inat

ion

of l

oca

lized

infe

ctio

n

and

sep

sis

Co

ulte

r

LH 7

50

Lee

et a

l. 20

13 [6

]El

der

lyV

olu

me

Co

nduc

tivi

ty

Scat

ter

(MA

LS)

148

±19.

3

150

±5.

3

134

± 13

.6

165

±24.

4

146

±6.4

130

±13

.2

<0.0

01

0.0

3

0.6

07

Dia

gno

sis

of s

epsi

s

Dis

crim

inat

ion

of l

oca

lized

infe

ctio

n

and

sep

sis

Co

ulte

r

DxH

80

0

Cel

ik e

t al

. 20

13 [4

5]N

eona

tes

Vo

lum

e

Co

nduc

tivi

ty

Scat

ter

148.

4 (1

1.1)

161.

5 (9

.1)

129

(10

.3)

170

.2 (

18.9

)

155.

9 (1

1.9)

125.

5 (1

1.8

<0.0

5

<0.0

5

<0.0

5

Wit

h C

RP /

IL-6

for

earl

y d

iagn

osi

s o

f

seps

is

Co

ulte

r

LH 7

80

Cha

ves

et a

l. 20

05

[46]

Ad

ults

Vo

lum

e

Co

nduc

tivi

ty

Scat

ter

143

±4.8

142

±2.6

146

±7.3

156

± 13

.5

141 ±

3.9

140

±10

.1

0.0

01

0.2

33

0.0

02

Dia

gno

sis

of a

cute

bac

teri

al in

fect

ion

Co

ulte

r

LH 7

50

Cha

rafe

dd

ine

et a

l. 20

11

[48]

Ad

ults

ND

W19

.8 (

1.1)

25.1

(3.6

)<0

.05

Dia

gno

sis

of S

IRS

Co

ulte

r

LH 7

50

Park

et

al. 2

011

[49]

Ad

ults

Vo

lum

e

Scat

ter

IG

153

±14.

3

141 ±

8.6

0.2

±0

.13

168

±28.

8

131 ±

14.4

2.4

±4.6

<0.0

5

<0.0

5

Dia

gno

sis

of s

epsi

sC

oul

ter

DxH

800

Sysm

ex X

E 21

00

Mak

kar

et a

l. 20

13 [5

1]N

eona

tes

I:M2

I:T2

53.1/

97.2

93.8

/94.

4

Dia

gno

sis

of s

epsi

sM

S 95

Sent

hiln

ayag

am e

t al

. 20

12

[52]

Ad

ults

IGC

2

IG (

%)2

86.3

/>90

92.2

/>90

Dia

gno

sis

of b

acte

rem

iaC

oul

ter

Act

Diff

5

Nig

ro e

t al

. 20

05

[53]

Neo

nate

sIG

(%

)233

/88

Pred

icti

on

of p

osi

tive

blo

od

cul

ture

Sysm

ex

XE-

210

0

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

142

8

Tab

le 2

. (co

ntin

ued

)

Art

icle

[re

f.]

Pop

ulat

ion

Para

met

ers

Co

ntr

ol1

Sep

tic1

P-va

lue

Co

rrel

atio

nA

nal

yzer

Park

et

al. 2

011

[54]

Neo

nate

sD

NI3

0 (

0-0

.1)2.

8 (0

.5-5

.3)

/ 16

.9 (

9.5-

35.6

)4

0.0

03

/

<0.0

01

Dia

gno

sis

of s

epsi

s an

d p

red

icti

on

of

seve

rity

Siem

ens

AD

VIA

212

0

Lim

et

al. 2

014

[55]

Ad

ults

DN

I548

.284

.20

.00

7Pr

edic

tio

n o

f sep

sis

in S

BP p

atie

nts

Siem

ens

Ad

via

2120

Nah

m e

t al

. 20

08

[56]

Ad

ults

DN

I619

.565

<0.0

5Pr

edic

tio

n o

f sep

sis

and

out

com

eSi

emen

s A

dvi

a

120

Lee

et a

l. 20

14 [5

7]N

eona

tes

DN

I70

.6 (

0.0

-2.1)

2.8

(0.8

0-5

.5)

<0.0

01

Pred

icti

on

of t

rue

bact

erem

iaSi

emen

s A

dvi

a

2120

Kim

et

al. 2

012

[58]

Ad

ults

DN

I81.9

6.2

0.0

05

Dia

gno

sis

of s

epsi

sSi

emen

s A

dvi

a

2120

Seo

k et

al.

2012

[59]

Neo

nate

sD

NI9

0.0

(0

.0-0

.0)

0.8

(0

.0-1

.7)

/ 3.

4 (1

.5-5

.3)

/18.

6 (9

.3-2

4.7)

0.0

00

1D

iagn

osi

s o

f sep

sis

Siem

ens

Ad

via

2120

IGC

= Im

mat

ure

gran

ulo

cyte

co

unt;

IG =

Imm

atur

e gr

anul

ocy

tes;

I:M

= Im

mat

ure

: Mat

ure

neut

roph

il ra

tio

; I:T

= Im

mat

ure

neut

roph

il : T

ota

l neu

tro

phil

rati

o; D

NI =

Del

ta N

eutr

oph

il In

dex

; MA

LS

= m

edia

n an

gle

light

sca

tter

; ND

W =

Neu

tro

phil

Dis

trib

utio

n W

idth

; CRP

= C

-rea

ctiv

e Pr

ote

in; S

IRS

= sy

stem

ic in

flam

mat

ory

res

po

nse

synd

rom

e; S

BP =

sp

ont

aneo

us b

acte

rial

per

ito

niti

s.

1 V

CS

dat

a pr

esen

ted

as

mea

n (r

ange

) o

r m

ean

± st

and

ard

dev

iati

on.

2 D

ata

pres

ente

d a

s se

nsit

ivit

y /

spec

ifici

ty fo

r pr

edic

tio

n o

f sep

sis

/ ba

cter

emia

(%

).3 D

ata

pres

ente

d a

s m

edia

n (r

ange

).4 D

ata

repr

esen

ts s

epsi

s /

sept

ic s

hock

.5 D

NI c

uto

ff v

alue

5.7

% (c

ont

rol =

< 5

.7%

and

sep

sis

= >

5.7%

); D

ata

pres

ente

d a

s %

occ

urre

nce

of s

epti

c sh

ock

am

ong

st S

PB p

atie

nts.

6 DN

I cut

off

val

ue 4

0%

(co

ntro

l = <

40%

and

sep

sis

= >4

0%

); D

ata

pres

ente

d a

s %

occ

urre

nce

of p

osi

tive

blo

od

cul

ture

.7 D

NI c

ont

rol =

blo

od

cul

ture

co

ntam

inat

ion;

DN

I sep

tic

in t

his

stud

y =

true

bac

tere

mia

. Dat

a pr

esen

ted

as

med

ian

(ran

ge).

8 DN

I co

ntro

l = s

urvi

vor;

DN

I sep

tic

= no

n su

rviv

or;

DN

I pre

sent

ed a

s m

edia

n (%

).9 D

ata

pres

ente

d a

s m

edia

n (r

ange

); S

epsi

s d

ata

pres

ente

d a

s SI

RS

/ Se

psis

/ S

ever

e Se

psis

.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

143

8

Standardized clinical flow cytometry

Multiparameter flow cytometry uses diffraction of light to measure cell size (forward scatter; FSC)

and cellular content (i.e., granularity; side scatter; SSC), along with laser based detection of cell

surface markers labeled by fluorescently labeled antibodies. Nowadays flow cytometry is broadly

used in the diagnosis of cancer, in assessing immune status of HIV patients, or for the monitoring of

immunological disease, but the use in diagnosis and monitoring of sepsis remains limited [60-63].

Recently, Roussel et al. applied a standardized clinical flow cytometric analysis of sepsis in 450

patients by analyzing their neutrophils [64,65]. They found that larger, less granular neutrophils

and increased numbers of immature neutrophils (identified with immunofluorescent staining of

cell surface markers) were associated with sepsis outcome and mortality.

Of note, several studies compared performance of measurement of the same parameters

between automated hematology analysers and flow cytometers and found overlapping results

[64-66]. Since the newest generations of automated hematology analyzers are able to measure

fluorescence equally well as flow cytometers, they can be used for the simultaneous measurement

of neutrophil morphological parameters and cell surface markers associated with their activation

status (e.g., CD11b, CD64) and neutrophil immaturity (e.g., CD16, CXCR2).

Automated Measurement of Neutrophil Deformability

Sepsis is associated with changes in neutrophil deformability that can be measured with different

assays [7,20-28,67,68]. For example, micropipette assays are useful for ex vivo determination

of deformability. With this method single neutrophils are aspirated against the tip of glass

micropipettes (usually with a lumen diameter of 2-5 μm and coated with plasma), followed

by time-lapse microscopic analysis of the neutrophils’ ability to migrate into the lumen. Other

approaches for measurement of deformability include the use of microchannels, magnetic

twisting cytometry and atomic force microscopy, which are currently being used in experimental

settings [67]. Although these methods are promising, their clinical use is not yet feasible, mostly

due to complexity of protocols and interpretation. A further detailed summary of developments in

these methods and their technical limitations can be found elsewhere [67].

Also, as mentioned above, techniques that measure deformability separate from other

variables are still absent. This was illustrated in samples of a small cohort of septic patients of which

blood transit time and number of obstructed microchannels were measured in an automated

multiple microchannel flow analyzer [68]. Whole blood from control patients passed easily through

the microchannels, while increased numbers of rigid leukocytes (predominantly neutrophils) in

blood from septic trauma patients caused significant obstruction of the flow, thereby increasing

both transit time and number of obstructed channels. These observations are also indicative of

changes in migration and chemotaxis, which are discussed in the next paragraph.

Automated Measurement of Neutrophil Migration and Chemotaxis

The activation state of neutrophils in response to septic conditions has an effect on their migration

and chemotaxis [5,6,8,9,33-35,68-76]. In particular, neutrophils from septic patients show less

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

144

8

migration and chemotactic responses when compared to neutrophils from healthy subjects. As

mentioned above, quantitative transwell assays are informative about total endpoint migration

and chemotaxis, but are not informative about the actual underlying dynamics. For that purpose,

parameters such as speed or direction of migration events are now being merged into clinical

use through the rapidly evolving and promising field of microfluidics [69]. Microfluidics in this

respect refers to methods in which small volumes of fluids (e.g., whole blood or isolated cells

in suspension) are applied to easy-to-use lab-on-a-chip microfluidic devices equipped with

standardized gradients of chemoattractants. These systems allow for fast assessment of neutrophil

migration and chemotaxis with arrayed time-lapse imaging and tracking algorithms [8,9,69-72]

(Table 3). For example, Berthier et al. tested their in house designed microfluidics device for

the analysis of neutrophils from an infant who presented with severe recurrent bacterial infections

due to a primary immunodeficiency disorder [71]. Their assay revealed impaired polarization and

chemotaxis in response to fMLP of neutrophils from the patient when compared to neutrophils

from its parents and an age-matched control. Using a similar approach, a clinical study in burn

patients (who usually present with a sepsis-like systemic inflammatory response syndrome)

observed an inverse relationship between neutrophil migration velocity towards a gradient of

fMLP and burn size in adults and children [9]. Interestingly, migration velocities were associated

with the development of post-burn bacteremia and sepsis, outlining the potential for clinical use

of such analytical approaches. Limitations to be dealt with include the necessity of isolation of

neutrophils from whole blood and gradient stabilization (such as described in reference 8).

A clinically more feasible approach, in which these limitations were minimized, was reported in

a prospective clinical study by Sackmann et al. to diagnose asthma [8]. Whole blood from patients

was pipetted into channels of a microfluidic device coated (or ‘primed’) with a neutrophil integrin

ligand (e.g., adhesion molecules such as E-selectin or ICAM-1), which resulted in direct capture of

neutrophils to the substrate. The unbound cells and other blood components were washed away

with laminar flow and the bound neutrophils were next allowed to migrate towards a controlled

gradient of fMLP already present in the device. Significantly lower neutrophil migration velocity

was predictive for the presence of asthma, with a sensitivity and specificity of 96% and 73%

respectively, and it discriminated asthma from allergic rhinitis. This assay required no additional

reagents and only 3 μL of whole blood, making it ideally suitable for situations in which only small

blood sample volumes can be acquired, such as in neonatal sepsis. Along with migration velocities

this assay can provide information on neutrophil polarization (i.e., leading edge formation) and

migration directionality through arrayed time-lapse imaging.

Although the direct use in sepsis diagnostics still remains to be investigated, microfluidic

devices provide a feasible, fast and cost-limiting aid. Additionally, microfluidic devices that

mimic the in vivo circumstances (e.g., with multiple chemokine gradients and substrates with,

or closely resembling, endothelial monolayers) are currently under development and could help

providing critical insight into neutrophil dynamics and underlying molecular cues in health and

disease [73-76].

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

145

8

Tab

le 3

. Stu

dies

mea

suri

ng n

eutr

oph

il m

igra

tio

n/ch

emo

taxi

s ve

loci

ty in

rel

atio

n to

infla

mm

atio

n an

d se

psis

Art

icle

[R

ef.]

Sett

ing

Mea

sure

men

tC

hem

oki

ne

Sub

stra

teC

on

tro

l1Pa

tien

ts1

P-va

lue

Dev

ice

Sack

man

n et

al.

2014

[5]

Clin

ical

: ast

hma

Che

mo

taxi

s

Vel

oci

ty

fMLP

(10

0 n

M)

P-se

lect

in (

100

μg/

mL)

1.6

1.33

0.0

02

K.O

.A.L

.A.

Butl

er e

t al

. 20

10 [6

]C

linic

al: b

urn

pati

ents

Che

mo

taxi

s

Vel

oci

ty

fMLP

(10

0 n

M)

Fibr

one

ctin

(10

0 μ

M)

18 ±

59

±6<0

.01

Mic

roflu

idic

dev

ice

Zho

u et

al.

200

4 [9

]Ex

per

imen

tal

Mig

rati

on

No

neG

lass

1.72

1.53 /3

.64 /

6.35

n.s.

/<0

.01/

<0.0

1

Flo

w c

ell s

yste

m

Dui

gnan

et

al. 1

986

[33]

Clin

ical

: po

st s

urgi

cal

seps

is

Che

mo

taxi

sC

asei

n (2

mg/

mL)

8μm

po

re M

embr

ane

86.8

±1.9

573

.4 ±

3.15

<0.0

2M

od

ified

Bo

yden

cham

ber

Chr

isto

u et

al.

1979

[34]

Clin

ical

: sep

sis

Che

mo

taxi

sC

asei

n

(5m

g/m

L)

8μm

po

re

Mem

bran

e

128.

1 ±2.

4590

.4 ±

2.95

<0.0

01

Mo

difi

ed B

oyd

en

cham

ber

Tave

res

et a

l. 20

02

[35]

Clin

ical

: sep

sis

Che

mo

taxi

sfM

LP (

100

nM

)8μ

m p

ore

Mem

bran

e

93.4

±6.

6651

±8.

36<0

.01

48 w

ell c

ham

ber

(Neu

ropr

ob

e)

Men

do

nça

et a

l. 20

05

[36]

Clin

ical

: bre

ast

canc

er

/ ba

cter

emia

Che

mo

taxi

sfM

LP (

100

nM

)8μ

m p

ore

Mem

bran

e

30.1

±8.3

62.

8 ±1

.36

<0.0

01

48 w

ell c

ham

ber

(Neu

ropr

ob

e)

Agr

awal

et

al. 2

00

8 [7

0]

Dev

ice

opt

imiz

atio

nC

hem

ota

xis

Vel

oci

ty

fMLP

, IL-

8V

ario

us17

No

neM

icro

fluid

ic d

evic

e

Bert

hier

et

al. 2

010

[71

]In

fant

wit

h re

curr

ent

infe

ctio

ns

Che

mo

taxi

s

Vel

oci

ty

fMLP

(10

0 n

M)

Fibr

one

ctin

0.15

80

.078

n =

1M

icro

fluid

ic d

evic

e

fMLP

= N

-fo

rmyl

met

hio

nine

-leu

cine

-phe

nyla

lani

ne; K

.O.A

.L.A

= k

it-o

n-a-

lid-a

ssay

.1 D

ata

pres

ente

d a

s m

ean

(ran

ge)

or

mea

n ±

stan

dar

d d

evia

tio

n (μ

m/m

in).

2 Neu

tro

phils

in H

BSS.

3 N

eutr

oph

ils in

HBS

S +

fMLP

(10

0nM

).

4 N

eutr

oph

ils in

HBS

S +

add

ed p

lasm

a pr

ote

ins.

5 Dat

a pr

esen

ted

as

mea

n d

ista

nce

± st

and

ard

err

or

of m

ean

(μm

).6 D

ata

pres

ente

d a

s m

ean

n o

f em

igra

ted

neu

tro

phils

± s

tand

ard

err

or.

7 Dat

a pr

esen

ted

for

chem

ota

xis

velo

city

of h

ealt

hy n

eutr

oph

ils to

war

ds

fMLP

gra

die

nt o

n P-

sele

ctin

sub

stra

te (

μm/m

in).

8 D

ata

pres

ente

d a

s m

ean

velo

city

(μm

/sec

).

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

146

8

CONCLUSIONS AND HYPOTHESISBased on our review of the literature we conclude that in sepsis the degree of inflammatory activation

drives changes in neutrophil morphology, mechanics and motility that are related to clinical

outcome. In particular, sepsis causes an increase in circulating numbers of larger, less granular,

more rigid neutrophils that show substantially diminished migration responses. The integration

of these changes into a diagnostic algorithm along with traditional biomarkers could facilitate

the identification of a clinically relevant hyperactivated septic neutrophil phenotype that can be

discriminated from other neutrophil phenotypes associated with local inflammation or resting

conditions (Figure 2). Until recently, technical limitations, such as the need for manual analysis of

the peripheral blood smear, still make made it difficult to properly evaluate the clinical potential of

measuring these changes in neutrophils. In fact, the traditional approach (i.e., assessing neutrophil

numbers and differentiation) has proven to be insufficient and there is a need for a more complete

identification of neutrophil phenotypes for sepsis diagnosis and clinical management [77]. For that

purpose, sophisticated automated hematology cell analysers and bedside microfluidic devices

reduce practical limitations and may provide feasible and time and cost-limiting aids.

The next step in validation of different phenotypes is to prospectively compare results

obtained by integrated measurements of these features in neutrophils between healthy subjects,

patients with local inflammation, and patients with sepsis. There are some challenges that need

to be considered before implementation of such an approach into the clinic is feasible. First, it is

becoming clear that neutrophils, instead of being one single cell type, are in fact a heterogeneous

group of cells with different fates and functions [78]. As such, neutrophils analyzed in peripheral

blood are not necessarily representative of the marginating pool in the microvasculature in vivo

that may have already depleted their innate immune functions (e.g., degranulation, migration,

NETosis). Second, diagnostic accuracy of the methodologies mentioned above needs to be

further investigated separately for proper establishment of clear cut-off values for each method

to discriminate healthy from septic neutrophils, also taking patient heterogeneity into account.

Third, the degree of changes in neutrophil morphology, mechanics and motility during sepsis

seems closely related to severity of disease and mortality of sepsis. However, how neutrophils

and these changes are exactly involved in sepsis pathophysiology, specifically in the development

of endothelial damage and subsequent organ failure, remains to be elucidated. To date there are

conflicting data on this latter issue. For example, some in vitro data imply increased adhesion of

septic neutrophils to the endothelium and loss of endothelial barrier function after incubation

with septic neutrophils [3,79]. In contrast, post mortem evaluation of human tissue after sepsis

revealed neutrophils in proximity of endothelium without evidence of endothelial damage or

increased accumulation of neutrophils in lung tissue, which was not associated with the extent

of damage resulting in acute lung injury (ALI) [80-82]. Identification of the here proposed more

complex neutrophil phenotypes may help in providing answers with regard to these questions

about the role of neutrophils in sepsis pathophysiology.

In conclusion, we propose that integration of neutrophil morphology, mechanics and motility

with these novel methods can lead to more accurate diagnosis, monitoring and prognosis of sepsis.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

147

8

Moreover, these analyses may be able to substantially contribute to the basic understanding of

sepsis, and in due time unveil new and specific treatment options as well as a means to determine

therapeutic effects of new treatments.

Acknowledgments

We acknowledge financial support from Tergooi Hospitals, the Drie Lichten foundation,

the Ter Meulen Fund (Royal Netherlands Academy of Arts and Sciences KNAW) and the IPRF Early

Investigators Exchange Program Award of the European Society for Pediatric Research (all to RZ).

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

148

8

Local InfectionHealthy Sepsis

Size

Granularity

Count

Immature:mature Ratio

Cell-Surface Molecules

Mature “Segmented” Neutrophil

Immature “Band” Neutrophil

Deformability

Migration

Delta Neutrophil Index

HIGH

x

Y

x

Y

x

Y

LOW

Figure 2. Schematic representation of healthy, local inflammatory and septic phenotypes of neutrophils.

Controlled inflammatory conditions drive changes in neutrophils. The excessive inflammatory conditions

of sepsis promote the presence of higher numbers of neutrophils, higher numbers of (larger) immature

neutrophils and large, less granular neutrophils in peripheral blood. Hypothetically, the average deformability

and migration velocities of septic neutrophils will be lower than under localized inflammatory conditions.

Novel methodologies can analyze many of these variables at the same time. Multiple morphological features

(size, volume, granularity, cell type and maturity) can be quickly assessed from high numbers of cells in small

volume blood samples with automated hematology analyzers and flow cytometry. The newest generation

of hematology analyzers can even incorporate measurement of critical cell surface molecules through

fluorescently labeled antibody based detection similar to flow cytometry. For neutrophil deformability

measurements, no clinically feasible technique has been developed yet. Neutrophil migration can be measured

at the bedside with microfluidic devices using small volumes of whole blood. Integrated measurement of

these variables with these novel methods can facilitate more accurate diagnosis, monitoring and management

of sepsis.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

149

8

REFERENCES1. Nauseef WM, Borregaard N. Neutrophils at

work. Nat Immunol 2014, 15(7):602-11.

2. King EG, Bauzá GJ, Mella JR, Remick DG.

Pathophysiologic mechanisms in septic shock.

Lab Invest 2014, 94(1):4-12.

3. Brown KA, Brain SD, Pearson JD, Edgeworth

JD, Lewis SM, Treacher DF. Neutrophils in

development of multiple organ failure in

sepsis. Lancet 2006, 368:157-69.

4. Celik IH, Demirel G, Aksoy HT, Erdeve O,

Tuncer E, Biyikli Z, Dilmen U. Automated

determination of neutrophil VCS parameters

in diagnosis and treatment efficacy of neonatal

sepsis. Pediatr Res 2012, 71(1):121-5.

5. Mardi D, Fwity B, Lobmann R, Ambrosch

A. Mean cell volume of neutrophils and

monocytes compared with C-reactive protein,

interleukin-6 and white blood cell count for

prediction of sepsis and nonsystemic bacterial

infections. Int J Lab Hematol 2010, 32(4):410-8.

6. Lee AJ, Kim SG. Mean cell volumes of

neutrophils and monocytes are promising

markers of sepsis in elderly patients. Blood

Res 2013, 48(3):193-7.

7. Skoutelis AT, Kaleridis V, Athanassiou GM,

Kokkinis KI, Missirlis YF, Bassaris HP. Neutrophil

deformability in patients with sepsis,

septic shock, and adult respiratory distress

syndrome. Crit Care Med 2000, 28(7):2355-9.

8. Sackmann EK, Berthier E, Schwantes EA,

Fichtinger PS, Evans MD, Dziadzio LL,

Huttenlocher A, Mathur SK, Beebe DJ.

Characterizing asthma from a drop of blood

using neutrophil chemotaxis. Proc Natl Acad

Sci USA 2014, 111(16):5813-8.

9. Butler KL, Ambravaneswaran V, Agrawal N,

Bilodeau M, Toner M, Tompkins RG, Fagan

S, Irimia D. Burn injury reduces neutrophil

directional migration speed in microfluidic

devices. PLoS One 2010, 30;5(7)

10. Kolaczkowska E, Kubes P. Neutrophil recruitment

and function in health and inflammation. Nat Rev

Immunol 2013, 13(3):159-75.

11. Newman TB, Draper D, Puopolo KM, Wi S,

Escobar GJ. Combining Immature and Total

Neutrophil Counts to Predict Early Onset Sepsis

in Term and Late Preterm Newborns: Use of

the I/T2. Pediatr Infect Dis J 2014, 33(8):798-802.

12. Van der Geest PJ, Mohseni M, Brouwer R, van

der Hoven B, Steyerberg EW, Groeneveld

AB. Immature granulocytes predict microbial

infection and its adverse sequelae in the intensive

care unit. J Crit Care 2014, 29(4):523-7.

13. Hoffstein ST, Friedman RS, Weissmann G.

Degranulation, membrane addition, and shape

change during chemotactic factor-induced

aggregation of human neutrophils. J Cell

Biol 1982, 95(1):234-41

14. Iba T, Hashiguchi N, Nagaoka I, Tabe Y,

Murai M. Neutrophil cell death in response

to infection and its relation to coagulation. J

Intens Care 2013, 1:13.

15. Fialkow L, Fochesatto Filho L, Bozzetti MC,

Milani AR, Rodrigues Filho EM, Ladniuk RM,

Pierozan P, de Moura RM, Prolla JC, Vachon E,

Downey GP. Neutrophil apoptosis: a marker of

disease severity in sepsis and sepsis-induced

acute respiratory distress syndrome. Crit

Care 2006, 10(6):R155.

16. McDonald B, Urrutia R, Yipp BG, Jenne CN, Kubes

P. Intravascular neutrophil extracellular traps

capture bacteria from the bloodstream during

sepsis. Cell Host Microbe 2012, 12(3):324-33.

17. Keshari RS, Jyoti A, Dubey M, Kothari N, Kohli

M, Bogra J, Barthwal MK, Dikshit M. Cytokines

induced neutrophil extracellular traps

formation: implication for the inflammatory

disease condition. PLoS One 2012, 7(10):e48111.

18. Hamaguchi S, Hirose T, Akeda Y, Matsumoto

N, Irisawa T, Seki M, Hosotsubo H, Tasaki O,

Oishi K, Shimazu T, Tomono K. Identification

of neutrophil extracellular traps in the blood of

patients with systemic inflammatory response

syndrome. J Int Med Res 2013, 41(1):162-8.

19. Hirose T, Hamaguchi S, Matsumoto N,

Irisawa T, Seki M, Tasaki O, Hosotsubo H,

Yamamoto N, Yamamoto K, Akeda Y, Oishi K,

Tomono K, Shimazu T. Presence of neutrophil

extracellular traps and citrullinated histone

H3 in the bloodstream of critically ill patients.

PLoS One 2014, 13;9(11):e111755.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

150

8

20. Pécsvárady Z1, Fisher TC, Fabók A, Coates

TD, Meiselman HJ. Kinetics of granulocyte

deformability following exposure to chemotactic

stimuli. Blood Cells 1992, 18(2):333-52.

21. Frank RS. Time-dependent alterations in

the deformability of human neutrophils

in response to chemotactic activation.

Blood 1990, 76(12):2606-12.

22. Worthen GS, Schwab B 3rd, Elson EL, Downey

GP. Mechanics of stimulated neutrophils: cell

stiffening induces retention in capillaries.

Science 1989, 245(4914):183-6.

23. Drost EM, Kassabian G, Meiselman HJ, Gelmont

D, Fisher TC. Increased rigidity and priming of

polymorphonuclear leukocytes in sepsis. Am J

Respir Crit Care Med 1999, 159(6):1696-702.

24. Erzurum SC, Downey GP, Doherty DE, Schwab

B 3rd, Elson EL, Worthen GS. Mechanisms

of lipopolysaccharide-induced neutrophil

retention. Relative contributions of adhesive

and cellular mechanical properties. J

Immunol 1992, 149(1):154-62.

25. Yoshida K, Ryoichi K, Wang Q, Doerschuk CM.

Neutrophil cytoskeletal rearrangements during

capillary sequestration in bacterial pneumonia in

rats. Am J Respir Crit Care Med 2006, 174:689-698.

26. Lichtman MA. Cellular deformability

during maturation of the myeloblast:

possible role in marrow egress. New Engl J

Med 1970, 283;260: 1124-1127.

27. Saito H, Lai J, Rogers R, Doerschuk CM. Mechanical

properties of rat bone marrow and circulating

neutrophils and their responses to inflammatory

mediators. Blood 2002, 99(6):2207-13.

28. Linderkamp O1, Ruef P, Brenner B, Gulbins E, Lang

F. Passive deformability of mature, immature,

and active neutrophils in healthy and septicemic

neonates. Pediatr Res 1998, 44(6):946-50.

29. Zonneveld R, Martinelli R, Shapiro NI, Kuijpers

TW, Plötz FB, Carman CV. Soluble adhesion

molecules as markers for sepsis and the potential

pathophysiological discrepancy in neonates,

children and adults. Crit Care 2014, 18;18(1):204.

30. Alves-Filho JC1, Spiller F, Cunha FQ. Neutrophil

paralysis in sepsis. Shock 2010, 34(1):15-21.

31. Reddy RC, Standiford TJ. Effects of sepsis

on neutrophil chemotaxis. Curr Opin

Hematol 2010, 17(1):18-24.

32. Alves-Filho JC, de Freitas A, Russo M, Cunha FQ.

Toll-like receptor 4 signalling leads to neutrophil

migration impairment in polymicrobial sepsis.

Crit Care Med 2006, 34(2):461-70.

33. Duignan JP, Collins PB, Johnson AH, Bouchier-

Hayes D. The association of impaired

neutrophil chemotaxis with postoperative

surgical sepsis. Br J Surg 1986, 73(3):238-40.

34. Christou NV, Meakins JL. Neutrophil function in

anergic surgical patients: neutrophil adherence

and chemotaxis. Ann Surg 1979, 190(5):557-64.

35. Tavares-Murta BM, Zaparoli M, Ferreira RB,

Silva-Vergara ML, Oliveira CH, Murta EF,

Ferreira SH, Cunha FQ. Failure of neutrophil

chemotactic function in septic patients. Crit

Care Med 2002, 30(5):1056-61.

36. Mendonça MA, Cunha FQ, Murta EF, Tavares-

Murta BM. Failure of neutrophil chemotactic

function in breast cancer patients treated

with chemotherapy. Cancer Chemother

Pharmacol 2006, 57(5):663-70.

37. Ahmed NA, McGill S, Yee J, Hu F, Michel RP,

Christou NV. Mechanisms for the diminished

neutrophil exudation to secondary

inflammatory sites in infected patients with

a systemic inflammatory response (sepsis).

Crit Care Med 1992, 7(11):2459-68.

38. Phillipson M, Heit B, Colarusso P, Liu L,

Ballantyne CM, Kubes P. Intraluminal crawling

of neutrophils to emigration sites: a molecularly

distinct process from adhesion in the recruitent

cascade. J Exp Med 2006, 203(12):2569-75.

39. Xu N, Lei X, Liu L. Tracking neutrophil

intraluminal crawling, transendothelial

migration and chemotaxis in tissue by intravital

video microscopy. J Vis Exp 2011, (55).

40. Lokuta MA, Nuzzi PA, Huttenlocher A. Analysis

of neutrophil polarization and chemotaxis.

Methods Mol Biol 2007, 412:211-29.

41. Zhou Y, Doerschuk CM, Anderson JM,

Marchant RE. Biomaterial surface-dependent

neutrophil mobility. J Biomed Mater

Res A 2004, 69(4):611-20.

42. Xiao W, Mindrinos MN, Seok J, Cuschieri J,

Cuenca AG, Gao H, Hayden DL, Hennessy

L, Moore EE, Minei JP, Bankey PE, Johnson

JL, Sperry J, Nathens AB, Billiar TR, West MA,

Brownstein BH, Mason PH, Baker HV, Finnerty

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

151

8

CC, Jeschke MG, López MC, Klein MB, Gamelli

RL, Gibran NS, Arnoldo B, Xu W, Zhang Y,

Calvano SE, McDonald-Smith GP, Schoenfeld

DA, Storey JD, Cobb JP, Warren HS, Moldawer

LL, Herndon DN, Lowry SF, Maier RV, Davis RW,

Tompkins RG; Inflammation and Host Response

to Injury Large-Scale Collaborative Research

Program. A genomic storm in critically injured

humans. J Exp Med 2011, 208(13):2581-90.

43. Tang BM, McLean AS, Dawes IW, Huang SJ, Lin

RC. Gene-expression profiling of peripheral

blood mononuclear cells in sepsis. Crit Care

Med 2009, 37(3):882-8.

44. Cornbleet PJ. Clinical utility of the band count.

Clin Lab Med 2002, 22(1):101-36.

45. Celik IH, Demirel G, Sukhachev D, Erdeve O,

Dilmen U. Neutrophil volume, conductivity and

scatter parameters with effective modeling of

molecular activity statistical program gives

better results in neonatal sepsis. Int J Lab

Hematol 2013, 35(1):82-7.

46. Chaves F, Tierno B, Xu D. Quantitative

determination of neutrophil VCS parameters by

the Coulter automated hematology analyzer:

new and reliable indicators for acute bacterial

infection. Am J Clin Pathol 2005, 124(3):440-4.

47. Bagdasaryan R, Zhou Z, Tierno B, Rosenman

D, Xu D. Neutrophil VCS parameters are

superior indicators for acute infection. Lab

Hematol 2007, 13(1):12-6.

48. Charafeddine KM, Youssef AM, Mahfouz

RA, Sarieddine DS, Daher RT. Comparison

of neutrophil volume distribution width

to C-reactive protein and procalcitonin as

a proposed new marker of acute infection.

Scand J Infect Dis 2011, 43(10):777-84.

49. Park DH, Park K, Park J, Park HH, Chae H, Lim

J, Oh EJ, Kim Y, Park YJ, Han K. Screening of

sepsis using leukocyte cell population data

from the Coulter automatic blood cell analyzer

DxH800. Int J Lab Hematol 2011, 33(4):391-9.

50. Bergen T, Steckhan D, Wittenberg T, Zerfass T.

Segmentation of leukocytes and erythrocytes

in blood smear images. Conf Proc IEEE Eng

Med Biol Soc 2008, 3075-8.

51. Makkar M, Gupta C, Pathak R, Garg S, Mahajan

NC. Performance evaluation of hematologic

scoring system in early diagnosis of neonatal

sepsis. J Clin Neonatol 2013, 2(1):25-9.

52. Senthilnayagam B, Kumar T, Sukumaran J, M J,

Rao K R. Automated measurement of immature

granulocytes: performance characteristics

and utility in routine clinical practice. Patholog

Res Int 2012, 483670.

53. Nigro KG, O’Riordan M, Molloy EJ, Walsh MC,

Sandhaus LM. Performance of an automated

immature granulocyte count as a predictor of

neonatal sepsis. Am J Clin Pathol 2005, 123(4):618-24.

54. Park BH, Kang YA, Park MS, Jung WJ, Lee SH,

Lee SK, Kim SY, Kim SK, Chang J, Jung JY, Kim

YS. Delta neutrophil index as an early marker

of disease severity in critically ill patients with

sepsis. BMC Infect Dis 2011, 11:299.

55. Lim TS, Kim BK, Lee JW, Lee YK, Chang S, Kim

SU, Kim do Y, Ahn SH, Han KH, Chon CY, Park JY.

Use of the delta neutrophil index as a prognostic

factor of mortality in patients with spontaneous

bacterial peritonitis: implications of a simple

and useful marker. PLoS One 2014, 9(1):e86884.

56. Nahm CH, Choi JW, Lee J. Delta neutrophil

index in automated immature granulocyte

counts for assessing disease severity of patients

with sepsis. Ann Clin Lab Sci 2008, 38(3):241-6.

57. Lee CH, Kim J, Park Y, Park YC, Kim Y, Yoon KJ, Uh Y,

Lee KA. Delta neutrophil index discriminates true

bacteremia from blood culture contamination.

Clin Chim Acta 2014, 427:11-4.

58. Kim HW, Ku S, Jeong SJ, Jin SJ, Han SH, Choi JY,

Kim JM, Song YG. Delta neutrophil index: could

it predict mortality in patients with bacteraemia?

Scand J Infect Dis 2012, 44(7):475-80.

59. Seok Y, Choi JR, Kim J, Kim YK, Lee J, Song

J, Kim SJ, Lee KA. Delta neutrophil index:

a promising diagnostic and prognostic marker

for sepsis. Shock 2012, 37(3):242-6.

60. Jaye DL, Bray RA, Gebel HM, Harris WA, Waller

EK. Translational applications of flow cytometry

in clinical practice. J Immunol 2012, 188(10):4715-9.

61. Monneret G, Venet F. Comment on

“Translational applications of flow cytometry in

clinical practice”. J Immunol 2012, 189(3):1099.

62. Umlauf VN, Dreschers S, Orlikowsky TW. Flow

cytometry in the detection of neonatal sepsis.

Int J Pediatr 2013, 2013:763191.

NO

VEL A

SPECTS O

F NEU

TROPH

ILS IN SEPSIS

152

8

63. Venet F, Lepape A, Monneret G. Clinical review: flow

cytometry perspectives in the ICU - from diagnosis

of infection to monitoring of injury-induced

immune dysfunctions. Crit Care 2011;15(5):231.

64. Roussel M, Benard C, Ly-Sunnaram B, Fest

T. Refining the white blood cell differential:

the first flow cytometry routine application.

Cytometry A 2010, 77(6):552-63.

65. Roussel M, Gros A, Gacouin A, Le Meur N, Le

Tulzo Y, Fest T. Toward new insights on the white

blood cell differential by flow cytometry:

a proof of concept study on the sepsis model.

Cytometry B Clin Cytom 2012, 82(6):345-52.

66. Groeneveld KM, Heeres M, Leenen LP, Huisman

A, Koenderman L. Immunophenotyping of

posttraumatic neutrophils on a routine haematology

analyser. Mediators Inflamm 2012, 509513.

67. Di Carlo D. A mechanical biomarker of cell state

in medicine. J Lab Autom 2012, 17(1):32-42.

68. Nishino M, Tanaka H, Ogura H, Inoue Y, Koh

T, Fujita K, Sugimoto H. Serial changes in

leukocyte deformability and whole blood

rheology in patients with sepsis or trauma. J

Trauma 2005, 59(6):1425-31.

69. Sackmann EK, Fulton AL, Beebe DJ. The present

and future role of microfluidics in biomedical

research. Nature 2014, 507(7491):181-9.

70. Agrawal N, Toner M, Irimia D. Neutrophil

migration assay from a drop of blood. Lab

Chip 2008, 8(12): 2054-61 .

71. Berthier E, Surfus J, Verbsky J, Huttenlocher A,

Beebe D. An arrayed high-content chemotaxis

assay for patient diagnosis. Integr Biol

(Camb) 2010, 2(11-12):630-8.

72. Jones CN, Hoang AN, Dimisko L, Hamza

B, Martel J, Irimia D. Microfluidic platform

for measuring neutrophil chemotaxis from

unprocessed whole blood. J Vis Exp 2014, (88).

73. Li Jeon N, Baskaran H, Dertinger SK, Whitesides

GM, Van de Water L, Toner M. Neutrophil

chemotaxis in linear and complex gradients

of interleukin-8 formed in a microfabricated

device. Nat Biotechnol 2002, 20(8):826-30.

74. Cho H, Hamza B, Wong EA, Irimia D. On-demand,

competing gradient arrays for neutrophil

chemotaxis. Lab Chip 2014, 14(5):972-8.

75. Myers DR, Sakurai Y, Tran R, Ahn B, Hardy

ET, Mannino R, Kita A, Tsai M, Lam WA.

Endothelialized microfluidics for studying

microvascular interactions in hematologic

diseases. J Vis Exp 2012, (64).

76. Kim D, Haynes CL. On-chip evaluation

of neutrophil activation and neutrophil-

endothelial cell interaction during neutrophil

chemotaxis. Anal Chem 2013, 85(22):10787-96.

77. Mare TA, Treacher DF, Shankar-Hari M,

Beale R, Lewis SM, Chambers DJ, Brown KA.

The diagnostic and prognostic significance

of monitoring blood levels of immature

neutrophils in patients with systemic

inflammation. Crit Care 2015, 25;19(1):57 .

78. Beyrau M, Bodkin JV, Nourshargh S.

Neutrophil heterogeneity in health and

disease: a revitalized avenue in inflammation

and immunity. Open Biol 2012, 2(11):120134.

79. Fox ED, Heffernan DS, Cioffi WG, Reichner JS.

Neutrophils from critically ill septic patients

mediate profound loss of endothelial barrier

integrity. Crit Care 2013, 17(5):R226.

80. Lucas S. The Autopsy Pathology of Sepsis-

Related Death, Severe Sepsis and Septic

Shock. Understanding a Serious Killer, Dr

Ricardo Fernandez (Ed.), ISBN: 978-953-307-

950-9, InTech, DOI: 10.5772/30095.

81. Kinoshita M, Mochizuki H, Ono S. Pulmonary

neutrophil accumulation following human

endotoxemia. Chest 1999, 116(6):1709-15.

82. Doerschuk CM. Mechanisms of leukocyte

sequestration in inflamed lungs.

Microcirculation 2001, 8(2):71-88.

9 Summary & Future Perspectives

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

159

9

This thesis focuses on early onset sepsis (EOS) amongst newborns in Suriname. In Part 1

demographics of newborns treated at the neonatal care facility in Suriname and the impact of

EOS are described, followed by the clinical dilemmas arising in the prediction of EOS in Part 2. In

Part 3 the vascular pathophysiology of EOS and potential for novel diagnostic methodologies are

described. This chapter summarizes and discusses the main findings of the various studies in this

thesis with implications for future research towards a novel diagnostic approach for EOS.

EPIDEMIOLOGY OF EARLY ONSET SEPSIS IN SURINAMETo date, detailed demographics of newborns in Suriname are absent. In 2008 the first referral

neonatal care facility in Suriname, with the ability for neonatal intensive care for newborns, opened

its doors at the Academic Hospital Paramaribo. In 2015, this facility transitioned to a modern

environment, along with implementation of interventions to improve neonatal care. In Chapter 2

of this thesis we evaluated the impact of this transition on mortality and morbidity of newborns.

We performed a retrospective study amongst 601 newborns admitted to the facility. We compared

outcomes of newborns between two 9-month periods before and after the transition in March

2015. After the transition more intensive care was delivered and more outborn newborns were

treated. Overall neonatal mortality rate of all inborn and outborn newborns was reduced from

23.4 to 13.4 deaths per 1,000 live births, along with a reduction in mortality of sepsis and asphyxia.

At the same time, mortality of newborns with a birth weight below 1,000 grams and incidence of

sepsis increased after the transition.

This study makes clear that two major challenges for future neonatal care in Suriname remain.

First, the reduction in neonatal mortality indicates a substantial improvement in the quality of

tertiary function of the facility and neonatal care in Suriname. However, despite these promising

results, it is important to realize that Suriname remains a developing country where political,

economic, and logistic challenges may negatively impact sustainability of this reduction in

mortality. We are currently designing a nationwide perinatal registry system and follow-up

epidemiological studies to monitor tertiary function, referral patterns, and morbidity and mortality

amongst Surinamese newborns. Second, incidence of sepsis increased after the transition of

the neonatal care facility. Although part of the increased sepsis incidence results from late onset

sepsis due to invasive lines and procedures, still half of all blood culture confirmed cases of sepsis

were cases of EOS. Additionally, over 30% of all admitted newborns were suspected of EOS and

empirically received antibiotics. Therefore, the main focus of this thesis was to summarize evaluate

clinical and pathophysiological aspects of EOS that may aid in improvement of early identification

and exclusion of EOS, with the final aim to initiate prompt treatment of infected newborns and

reduce unnecessary antibiotic usage amongst uninfected ones.

PREDICTION OF EARLY ONSET SEPSISPrediction of EOS is complicated due to the fact that clinical symptoms and inflammatory

biomarkers are often not specific for presence of EOS [1]. Like in many Western countries, clinical

protocol at the neonatal care facility in Suriname prescribes a combination of perinatal risk factors,

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

160

9

clinical symptoms of the newborn, and serial assessment of inflammatory biomarkers C-reactive

protein (CRP) and white blood cell count, to predict presence or absence of EOS within 72 hours

after birth. In Part 2 of this thesis we describe tools that may be of additive value in the prediction

of EOS in the clinic.

The online available EOS calculator is a novel tool to predict EOS and help decision making

on start and duration of antibiotic treatment [2]. The EOS calculator is based on five objective

maternal parameters and clinical evaluation of the newborn straight after birth, and provides a risk

estimate on EOS. It is unclear how the EOS calculator relates to levels of CRP and leukocyte and

thrombocyte counts in the first 72 hours of life. Increase of CRP and leukopenia have been shown

to be associated with blood culture positive EOS. In Chapter 3 we investigated the hypothesis

that higher EOS calculator results are associated with increase in CRP within 24-48 hours and low

leukocyte counts. EOS risk estimates were calculated for 108 newborns of 34 weeks of gestational

age, in whom antibiotics were started for suspected EOS. EOS risk estimates were retrospectively

compared to infection parameters CRP, and leukocyte and trombocyte counts. In contrast to our

hypothesis, high EOS risk at birth was consistently correlated with lower CRP and leukocyte counts

within 24 hours after the start of antibiotics, but not with infection parameters after 24 hours.

The study in Chapter 3 does not show correlation with infection parameters CRP and leukocyte

counts that are currently commonly used in the clinic to help decision making on start and duration

of antibiotic treatment in the clinic. However, in a large recent study amongst 204,485 newborns

in the United States determination of EOS risk with the EOS calculator reduced the number of

newborns that received laboratory testing and empirical antibiotic treatment [3]. Retrospective

analysis of the Dutch cohort used in Chapter 3 showed that application of the EOS calculator could

reduce antibiotic treatment with 50% [4]. To further enhance its clinical utility, it may be useful to

investigate association of the EOS calculator with biomarkers, such as immature granulocytes and

markers of endothelial cell activation, discussed in this thesis. Integration of the EOS calculator

with these biomarkers into a novel diagnostic approach is proposed in the last paragraph of

this chapter.

In Chapter 4 we investigated another option to predict EOS and to help decisions on start

and duration of antibiotic treatment. Automated measurement of immature-to-total-granulocyte

(I/T) has been shown to have negative predictive value of EOS [5]. We retrospectively evaluated

a one-point measurement of immature-to-total-granulocyte (I/T) ratio in predicting duration of

antibiotic treatment in EOS in a cohort of Surinamese newborns. I/T ratio was lower in newborns

in whom antibiotics were discontinued at 48-72 hours after start after which they all remained

healthy. We conclude that low I/T ratio may help to increase the threshold to start empirical

antibiotic treatment or to guide safe stoppage of antibiotics after 48-72 hours. Further prospective

investigations in larger cohorts of newborns are necessary to evaluate clinical utility of a one-point

measurement of I/T ratios, and to establish appropriate cut-off values. Nonetheless, it is a quickly

available measurement (i.e., within 10 minutes after blood draw) to help decision-making on start

of antibiotics in suspected EOS. Additionally, the fact that automated hematology analyzers are

becoming universally available in the non-Western world favors their implementation there.

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

161

9

THE VASCULAR PATHOPHYSIOLOGY OF EARLY ONSET SEPSIS The dilemmas in prediction of EOS in the clinic occur because the pathophysiology of EOS is

poorly understood. In Part 3 of this thesis we focus on the vascular pathophysiology of EOS and

the potential of its molecular aspects for translation into novel diagnostic methodologies.

The vascular pathophysiology of sepsis is associated with interactions between leukocytes

and the vascular endothelium [6]. During sepsis, adhesion molecules are expressed on the cell

membranes of both cell types that orchestrate leukocyte rolling on, adhesion to, and transmigration

across the endothelium [7]. As inflammation progresses, adhesion molecules accumulate in

the blood as soluble forms after shedding by shedding enzymes, or ‘sheddases’, such as matrix

metalloproteinase-9 (MMP-9) and neutrophil elastase [8]. In Chapter 6 we review the studies that

have tested the predictive value of soluble adhesion molecules (sCAMs) in sepsis pathophysiology

in newborns, children and adults. Four endothelial sCAMs, specifically P-selectin, E-selectin,

vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1), along

with one leukocyte adhesion molecule, namely L-selectin, were associated with sepsis. While

increased levels of these sCAMs generally correlated well with the presence of sepsis, their degree

of elevation was poorly predictive of sepsis severity scores, outcome and mortality. Separate

analyses of newborns, children, and adults demonstrated significant age-dependent differences

in both basal and septic levels of sCAMs. Based on the results reported in this review, we proposed

two novel directions for improving clinical utility of sCAMs: 1) the combined simultaneous analysis

of levels of soluble adhesion molecules and their sheddases, and 2) taking age into account in

the interpretation of their levels.

In Chapter 7 we applied this approach in a Surinamese cohort of 20 healthy newborns and 71

newborns with suspected EOS, included within 72 hours after birth. We hypothesized that sCAMs

and sheddases circulate at higher levels in blood culture positive EOS in newborns and that they

are useful as biomarkers for EOS. Soluble CAMs sP-selectin, sE-selectin, sVCAM-1, sICAM-1, and

platelet and endothelial cell adhesion molecule-1 (sPECAM-1), sheddases MMP-9 and neutrophil

elastase, and sheddase antagonist tissue-inhibitor of metalloproteinases-1 (TIMP-1) were measured

simultaneously in serum of 91 newborns. Of the newborns, six (8.5%) had a positive blood culture.

At start of antibiotic treatment and after 48-72 hours no differences were found in levels of sCAMs

and sheddases between blood culture positive EOS, blood culture negative EOS and controls. In

contrast to our hypothesis, these data show that endothelial CAM shedding was not increased in

EOS. Additionally, levels of sCAMs and sheddases were similar in all newborns between straight

and six days after birth, indicating that sCAM shedding remains unchanged in early newborn life.

Levels of sCAMs found in our study corresponded well with levels in other studies in newborns.

However, when compared with earlier reports in adults sCAM levels in newborns are higher. We

concluded that other mechanisms, such as perinatal stress during birth, may drive overall high

levels in all newborns which precludes discrimination between septic and healthy newborns based

on these levels. For these reasons, sCAMs and sheddases may not prove to be useful as biomarkers

for EOS. Thus, our study indicates that simultaneous measurement of sCAMs and sheddases, as

proposed in our review in Chapter 6, does not provide a satisfactory improvement of clinical utility

of sCAM levels in prediction of EOS.

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

162

9

Vascular inflammation and leakage in sepsis is mediated by Angiopoietin (Ang)-1 and Ang-2

and their binding associated phosphorylation of the endothelial Tie-2 receptor. Levels of Ang-2

change in adults and children during sepsis, which is associated with severity of sepsis. In Chapter 8

we investigated serum levels of Ang-1 and Ang-2 in newborns within 72 hours after birth.

A prospective study was performed in the same cohort of newborns as described in Chapter 7.

At start of antibiotic treatment Ang-1 serum levels were significantly lower and Ang-2 and Ang-2/

Ang-1 serum protein ratios higher in newborns with blood culture positive EOS than in blood

culture negative EOS and controls. These levels were not dependent on timing of first blood

draw after birth. After 48-72 hours, levels of Ang-1 further decreased in blood culture positive

EOS, while in the other groups no change was observed. These findings support the hypothesis

that a dysbalance in the Angiopoietins is associated with the (vascular) pathophysiology of EOS.

Additionally, these findings suggest potential for the Angiopoietins as biomarkers for EOS.

We propose further investigations into the Angiopoietin in EOS at two complementary levels.

First, we propose to measure Ang-1 and Ang-2 levels in peripheral and cord blood in a mouse

model of pregnancy to study both maternal and perinatal factors that may influence Ang-1 and

Ang-2 levels in neonatal mice [9]. In addition, inoculation of neonatal offspring born in this study

with Group B Streptococcus (GBS), to model EOS, within 72 hours could further reveal the vascular

pathophysiology of EOS [10]. Second, from the results of our first study we hypothesize that

serial measurement of high Ang-1, low Ang-2, and low Ang-2/Ang-1 ratio may negatively predict

presence of EOS, and thus be extra arguments for safe stoppage of antibiotics. We propose further

evaluation of Ang-1 and Ang-2 as biomarkers for EOS by repeating the study in Chapter 8 in a larger

Surinamese cohort to establish sensitivity, specificity and appropriate cut-off and predictive values

of the Angiopoietins for EOS. Maternal and perinatal factors may influence levels of Ang-1, Ang-2

and Ang-2/Ang-1 ratios in newborns, for which logistic regression analysis should be performed.

Additionally, the methodological possibilities to enable quick measurement of Ang-1 and Ang-2

levels for clinical purpose should be explored.

While the results from earlier chapters in this thesis provide new avenues for identification and

exclusion of EOS, a continuing need for (development of) novel and clinically useful diagnostic

tools remains. Alterations in neutrophil morphology (size, shape and composition), mechanics

(deformability), and motility (chemotaxis and migration) have been observed during sepsis.

In Chapter 9 we summarized features of neutrophil morphology, mechanics, and motility that

change during sepsis and combined that with an investigation into their clinical utility as markers

for sepsis through measurement with existing and novel technologies. When compared to resting

conditions, sepsis causes an increase in circulating numbers of larger, more rigid neutrophils

that show diminished granularity, migration and chemotaxis. Combined measurement of these

parameters may provide a more complete view on neutrophil phenotype manifestation. For that

purpose, sophisticated automated hematology analysers, microscopy and bedside microfluidic

devices provide clinically feasible, high throughput, and cost limiting means. We propose that

integration of features of neutrophil morphology, mechanics and motility with these new analytical

methods can be useful as markers for diagnosis, prognosis and monitoring of sepsis, and may even

contribute to our basic understanding of its pathophysiology (See also Appendix I).

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

163

9

OPTIONS AND CHALLENGES FOR A NOVEL DIAGNOSTIC APPROACH FOR EARLY ONSET SEPSIS As discussed above, the major challenge in EOS is its timely identification for prompt initiation of

antibiotic treatment, while preventing unnecessary antibiotic treatment in uninfected newborns.

This is especially true for low resource settings, such as Suriname, where the risk for EOS is relatively

high, and the threshold to start empirical antibiotic treatment is relatively low, when compared to

Western countries. To date, only serial measurement of low levels of CRP, combined with a negative

blood culture and clinically improved newborn, showed negative predictive value for EOS [11]. In

a recent randomized controlled trial amongst 1,710 newborns in eighteen clinics in four European

countries and Canada, a clinical decision-making regimen, in which a four point measurement of

procalcitonin was added to the standard regimen of measurement of CRP and white blood cell

counts alone, was superior in reducing duration of antibiotic treatment [12]. Data in this thesis

show that incidence of blood culture positive EOS in Suriname is higher than reported in this trial

(i.e., estimated between 8-10% versus 1-2%, respectively) indicating that newborns in Suriname

are at higher risk for EOS. Additionally, the safety nets and facilities to properly follow-up clinical

evolution of sent home newborns in whom antibiotics are discontinued, such as home maternity

care, are absent in Suriname. For these reasons, the reported superior new regimen including

procalcitonin cannot be safely applied directly to the Surinamese situation. However, the standard

regimen described in this study was similar to the regimen that is currently used in Suriname.

Thus, it would be interesting to perform a similar randomized controlled ‘non-inferiority’ trial to

compare a novel diagnostic approach with the standard regimen in Suriname.

Starting point of this proposed novel diagnostic approach is the EOS calculator. Since the EOS

calculator is freely available online (also as an app for a smartphone), it is the most affordable

tool to guide clinicians in decision-making on start of antibiotics in cases of suspected EOS in

low resource settings such as Suriname. Especially at medical posts in rural areas of Suriname,

the EOS calculator is an easy-to-use tool to help decision-making on transport of newborns

to the neonatal care facility in the capital Paramaribo for evaluation and treatment. Before

implementation in Suriname, it is important to realize that for optimal performance the EOS

calculator algorithm uses local incidence of EOS is needed. Another variable in the algorithm is

maternal GBS colonization status, which is usually unknown in pregnant women in the Surinamese

setting. Moreover, our prospective studies showed that only gram-negative EOS occurred in our

cohort, indicating that GBS colonization may play an inferior role as causative pathogen of EOS

in Suriname. We are currently performing a prospective observational cohort study in pregnant

women and their newborns in Suriname to evaluate the exact incidence and local risk factors

for EOS and performance of the current EOS calculator. This is intended to ultimately create

a customized EOS calculator with variables that are specific for Suriname. Further improvement

of EOS calculator performance can be achieved when used in combination with novel biomarkers

discussed in this thesis. As discussed above, appropriate cut-off values for both I/T ratio and levels

of Ang-1 and Ang-2 have to be established in larger prospective studies. Once these have been

established, in Suriname the novel diagnostic approach could consists of the following if a newborn

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

164

9

presents with suspected EOS: 1) immediate determination of EOS risk with a Suriname-specific

EOS calculator, combined with 2) immediate measurement of I/T ratio to help decision-making

on whether to start antibiotic treatment, and then, if antibiotics are started, 3) repeated Ang-2/

Ang-1 ratio determination to help decide whether to continue antibiotic treatment after 48-72

hours after start. This novel diagnostic approach could reduce antibiotic treatment in suspected

EOS in two complementary ways, namely the prevention of start of unnecessary antibiotics, and

safe stoppage of antibiotic treatment after 48-72 hours in the high-risk low resource setting. If

successful, both scenarios would be a major step forward in reducing the burden of antibiotic

treatment in newborns in Suriname and similar low resource settings.

A FUTURE CASE OF SUSPECTED EARLY ONSET SEPSIS IN SURINAME IN 2030After a boat ride from her village down the Suriname River, the mother arrives at the nearest mission

post in Debike1. She is pregnant for eight months and her water has broken a few days earlier. She

has korsu2. The datra3 at the mission post uses an application on his smartphone and puts in her

temperature. He explains that the result of the app tells him to send her to the new hospital in

the city of Paramaribo to give birth because her unborn child may have an infection. She gives

birth to a daughter there the next day. The doctors take her to the baby ward and take her blood.

The results of the blood test come back within 10 minutes, after which they start antibiotics. After

two days the baby is healthy and feeding well. The doctors take her blood again. The nurse tells her

the results are fine and that they will stop the antibiotic treatment. The next day the mother rides

the boat home to her village with a healthy daughter.

1 Village located along the Suriname River in the district Sipaliwini in the interior of Suriname

Translated from the Surinamese language (Sranan Tongo):2 physician;3 fever.

SUM

MA

RY A

ND

FUTU

RE PERSPEC

TIVES

165

9

REFERENCES1. van Herk W, Stocker M, van Rossum AMC.

Recognising early onset neonatal sepsis: An

essential step in appropriate antimicrobial use.

J Infect 2016, 72:S77–82.

2. Escobar GJ, Puopolo KM, Wi S, Turk BJ,

Kuzniewicz MW, Walsh EM, Newman

TB, Zupancic J, Lieberman E, Draper

D. Stratification of risk of early-onset

sepsis in newborns ≥ 34 weeks’ gestation.

Pediatrics 2014, 133(1):30–6.

3. Kuzniewicz MW, Puopolo KM, Fischer A,

Walsh EM, Li S, Newman TB,, Kipnis P, Escobar

GJ. A Quantitative, Risk-Based Approach to

the Management of Neonatal Early-Onset

Sepsis. JAMA Pediatr 2017, 171(4):365-371.

4. Kerste M, Corver J, Sonnevelt MC, van Brakel

M, van der Linden PD, M Braams-Lisman BA,

Plötz FB. Application of sepsis calculator in

newborns with suspected infection. J Matern

Fetal Neonatal Med 2016, 29(23):3860-5.

5. Mikhael M, Brown LS, Rosenfeld CR. Serial

neutrophil values facilitate predicting

the absence of neonatal early-onset sepsis. J

Pediatr 2014, 164(3):522-8

6. Aird WC. The role of the endothelium in

severe sepsis and multiple organ dysfunction

syndrome. Blood 2003, 101(10):3765-77.

7. Ley K, Laudanna C, Cybulsky MI, Nourshargh

S. Getting to the site of inflammation:

the leukocyte adhesion cascade updated. Nat

Rev Immunol 2007, 7(9):678-89.

8. Garton KJ, Gough PJ, Raines EW. Emerging roles

for ectodomain shedding in the regulation

of inflammatory responses. J Leukoc

Biol 2006, 79(6):1105-16.

9. Reynolds LP, Borowicz PP, Vonnahme KA,

Johnson ML, Grazul-Bilska AT, Wallace JM, Caton

JS, Redmer DA. Animal models of placental

angiogenesis. Placenta 26(10):689-708, 2005.

10. Mancuso G, Midiri A, Beninati C, Biondo

C, Galbo R, Akira S, Henneke P, Golenbock

D, Teti G. Dual role of TLR2 and myeloid

differentiation factor 88 in a mouse model

of invasive Group B streptococcal disease. J

Immunol 2004, 172: 6324-6329.

11. Hofer N, Zacharias E, Müller W, Resch B. An

update on the use of C-reactive protein in

early-Onset neonatal sepsis: Current insights

and new tasks. Neonatology 2012;102:25–36.

12. Stocker M, van Herk W, El Helou S, Dutta

S, Fontana MS, Schuerman FABA, van den

Tooren-de Groot RK, Wieringa JW, Janota J, van

der Meer-Kappelle LH, Moonen R, Sie SD, de

Vries E, Donker AE, Zimmerman U, Schlapbach

LJ, de Mol AC, Hoffman-Haringsma A, Roy M,

Tomaske M, Kornelisse RF, van Gijsel J, Visser

EG, Willemsen SP, van Rossum AMC; NeoPInS

Study Group. Procalcitonin-guided decision

making for duration of antibiotic therapy in

neonates with suspected early-onset sepsis:

a multicentre, randomised controlled trial

(NeoPIns). Lancet 2017, pii. 6736(17)31444-7.

& Appendices

APPEN

DIX

I

171

&

APPENDIX I: MEASUREMENT OF FUNCTIONAL AND MORPHODYNAMIC NEUTROPHIL PHENOTYPES IN SYSTEMIC INFLAMMATION AND SEPSIS

Rens Zonneveld, Grietje Molema, Frans B. Plötz

Letter to the editor of Critical Care

Critical Care 2016, 20:235-36.

We read with great interest the review by Leliefeld et al. [1] published in Critical Care, on the role

of neutrophils in immune paralysis during systemic inflammation (SI). This is of high clinical

importance since immune paralysis potentially increases susceptibility to new infections or may

cause inability to clear existing infections leading to detrimental outcome. One mechanism

proposed for immune paralysis is the release of neutrophil populations with decreased microbicidal

properties [1]. During SI heterogeneous subsets of neutrophils exist with different priming states

and functions [2]. In particular, large numbers of immature neutrophils appear in the circulation

with diminished expression of receptors, important for pathogen killing [3]. We recently reviewed

literature on morphodynamic changes in neutrophils during sepsis [4]. Sepsis increases their

circulating numbers (and percentages of immature neutrophils) and cell size and stiffness, and

decreases migration/chemotaxis, compared to non-diseased conditions or mild infection. Septic

neutrophils are also prone to produce neutrophil extracellular traps (NETs).

Both reviews outline shifting neutrophil identity from an innate responder to a complex immune

cell with different functional and morphodynamic phenotypes depending on the underlying

pathology. The relationship between changes in functional and morphodynamic phenotypes

is not completely understood. For example, inability of circulating neutrophils to migrate to

infectious sites during SI may be the result of diminished expression of chemotactic receptors

(e.g., CXCR2), which is most profound in newly released immature granulocytes [1]. Others have

suggested a hyperadhesive state to the endothelium, due to overexpression of integrins (e.g.,

CD11b/CD18) on their membrane, thereby decreasing their ability to migrate [5]. Whether this

impaired migration interferes with pathogen killing capacity needs to be investigated.

It is important to know that detecting morphodynamic changes in neutrophils is nowadays

clinically feasible with novel technologies [4]. Automated hematology analysers can measure

(immature) neutrophil counts and cell sizes, while microfluidic devices can measure migration

velocities. These methods represent a diagnostic toolbox that enables multi-parameter analysis

of neutrophils during SI and sepsis. When combined with technologies that are still in their infancy

(e.g., flowcytometry to measure degranulation), bedside measurement of functions of neutrophils

and in vitro studies as proposed by Leliefeld (e.g., measurement of intra-or extracellular killing

activity) will become feasible. This approach will combine functional and morphodynamic

neutrophil phenotypes into clinical settings for better understanding of the exact role of

neutrophils in SI and sepsis. Ultimately, this may help to determine whether neutrophils can be

considered rational targets for therapy.

APPEN

DIX

I

172

&

DECLARATIONSEthical approval and consent to participate

Not applicable

Consent for publication

Not applicable

Availability of supporting data

Not applicable

Competing interests

The authors declare that they have no competing interests

Funding

RZ was sponsored by the Thrasher Research Fund

Author’s contributions

RZ, GM and FBP conceived, drafted and finalized the manuscript

Acknowledgments

Not applicable

Authors’ information

Not applicable

APPEN

DIX

I

173

&

REFERENCES1. Leliefeld PH, Wessels CM, Leenen LP,

Koenderman L, Pillay J. The role of neutrophils

in immune dysfunction during severe

inflammation. Crit Care 2016, 20(1):73.

2. Pillay J, Ramakers BP, Kamp VM, Loi ALT,

Lam SW, Hietbrink F, et al. Functional

heterogeneity and differential priming of

circulating neutrophils in human experimental

endotoxemia. J Leukoc Biol 2010, 88:211–20.

3. Navarini AA, Lang KS, Verschoor A, Recher M,

Zinkernagel AS, Nizet V, et al. Innate immune-

induced depletion of bone marrow neutrophils

aggravates systemic bacterial infections. Proc

Natl Acad Sci U S A 2009, 106:7107–12.

4. Zonneveld R, Molema G, Plötz FB. Analyzing

Neutrophil Morphology, Mechanics, and

Motility in Sepsis: Options and Challenges

for Novel Bedside Technologies. Crit Care

Med 2016, 44(1):218-28.

5. Blom C, Deller BL, Fraser DD, Patterson EK,

Martin CM, Young B, Liaw PC, Yazdan-Ashoori

P, Ortiz A, Webb B, Kilmer G, Carter DE,

Cepinskas G. Human severe sepsis cytokine

mixture increases β2-integrin-dependent

polymorphonuclear leukocyte adhesion to

cerebral microvascular endothelial cells in

vitro. Crit Care 2015, 19:149.

APPEN

DIX

II

174

&

APPENDIX II: SAMENVATTING (SUMMARY IN DUTCH)Early Onset Sepsis (EOS) wordt gedefinieerd als een ernstige bacteriële infectie in de bloedbaan

van een pasgeborene die zich manifesteert binnen 72 uur na geboorte. In Westerse landen

krijgt 1 op de 1000 (0.1%) pasgeborenen EOS. Het aantal gevallen van EOS in Suriname is niet

bekend, maar ligt waarschijnlijk veel hoger. Tijdens EOS ontstaat een ontstekingsreactie in

de bloedvaten waarbij met name afweercellen (zogenaamde witte bloedcellen) en het endotheel -

de binnenbekleding van bloedvaten en de barrière tussen bloed en de weefsels - betrokken

zijn. Tijdens EOS kan door verlies van de barrièrefunctie van het endotheel vocht in de weefsels

uittreden wat leidt tot ernstige problemen met de ademhaling en een lage bloeddruk. Het tijdig

starten van antibiotica kan sterfte als gevolg van EOS voorkomen. Echter, EOS kan alleen worden

bevestigd met een kweek van het bloed die pas na enkele dagen groei van aanwezige bacteriën

laat zien. Daarnaast zijn symptomen van EOS slecht te onderscheiden van symptomen die elke

pasgeborene kan vertonen als gevolg van aanpassingsproblemen door de geboorte. Hierdoor is

het tijdig aantonen of uitsluiten van EOS niet goed mogelijk en worden veel pasgeborenen te laat

of juist onnodig behandeld met antibiotica.

Het onderzoek in dit proefschrift richt zich op EOS in Suriname en op aspecten van

de ontstekingsreactie in het bloedvat en daarop gebaseerde methoden van detectie die bij

kunnen dragen aan het stellen van de juiste diagnose. In Suriname wordt gespecialiseerde

zorg aan pasgeborenen geleverd in de Neonatal Care Facility van het Academisch Ziekenhuis

Paramaribo. Het eerste deel van dit proefschrift beschrijft hoe vaak EOS onder de daar

behandelde pasgeborenen voorkomt. In het tweede deel worden twee reeds beschikbare

en toegankelijke methoden getoetst als mogelijk diagnostische test voor EOS in Suriname.

Dit betreft een online verkrijgbaar risicomodel voor EOS, de zogenaamde EOS Calculator, en

de geautomatiseerde kwantitatieve meting van een bepaald type afweercel, de immature

granulocyt. In het derde deel worden drie groepen circulerende eiwitten gemeten in het bloed van

Surinaamse pasgeborenen. Dit zijn de endothelial cell adhesion molecules (CAMs) en sheddases,

die samen de interactie tussen afweercellen en het endotheel verzorgen, en de Angiopoietins, die

betrokken zijn bij het onderhouden van de barrièrefunctie van het endotheel tijdens ontsteking.

Voor deze studies stelden we ons de vraag of deze eiwitten een voorspellende waarde hebben

voor het beloop van de bacteriële infectie.

De resultaten van het onderzoek zijn als volgt. Na vernieuwing van de Neonatal Care Facility

in Paramaribo blijft EOS een groot probleem. Het aantal gevallen van met een bloedkweek

bewezen EOS na de vernieuwing was nog steeds tussen de 50-100 (5-10%) op 1000 pasgeborenen,

hetgeen vele malen hoger is dan in de Westerse landen. 30% van de pasgeborenen kreeg

antibiotica voor een verdenking op EOS. De EOS Calculator helpt om het risico op EOS in te

schatten. Lage bloedwaarden van de immature granulocyt waren voorspellend voor afwezigheid

van EOS en kunnen helpen om onnodige behandeling met antibiotica te voorkomen of de duur

van de behandeling te verkorten. De bloedspiegels van de CAMs en sheddases waren hoog maar

niet geassocieerd met het voorkomen van EOS. Waarschijnlijk hebben deze hoge waarden een

andere oorzaak dan EOS, zoals stress omtrent de geboorte. Meting van de Angiopoietins in

APPEN

DIX

II

175

&

dezelfde groep Surinaamse pasgeborenen liet zien dat er een disbalans ontstaat tijdens EOS tussen

de twee belangrijke vormen Ang-1 en Ang-2. Ang-1 bloedwaarden zijn lager en Ang-2 waarden

hoger in pasgeborenen met EOS waardoor de Ang-2/Ang-1 ratio verandert. Deze bevindingen

geven aan dat het endotheel wordt geactiveerd en betrokken is bij de ontstekingsreactie in

de bloedvaten tijdens EOS. Verder onderzoek moet uitwijzen of het meten van deze eiwitten ook

kan worden gebruikt om EOS aan te tonen of uit te sluiten.

De bevindingen van dit proefschrift laten zien dat EOS een groot probleem is in Suriname.

De combinatie van risico inschatting op EOS met de EOS calculator, mits aangepast voor

de Surinaamse situatie, meting van de immature granulocyt en seriële meting van de Angs kan in

de toekomst mogelijk helpen om onnodig gebruik van antibiotica te reduceren.

APPEN

DIX

III

176

&

APPENDIX III: DANKWOORDHet onderzoek in dit proefschrift werd uitgevoerd op drie verschillende continenten en bracht

mij van de kinderafdeling van Tergooi Ziekenhuizen in Blaricum naar het laboratorium van Harvard

Medical School in Boston, en vervolgens van de kinderafdeling van het Academisch Ziekenhuis

Paramaribo naar het laboratorium van het Universitair Medisch Centrum in Groningen. Ik heb

de eer gehad om samen te werken met een groot aantal zeer inspirerende mensen door wie dit

proefschrift tot stand is gekomen.

Allereerst, copromotor en mentor dr. Frans Plötz. Beste Frans, man van het eerste uur van al het

werk in dit proefschrift. In de afgelopen vijf jaar wil ik graag zeggen dat ik een vriend rijker ben

geworden. Ik kan me onze eerste ontmoeting nog herinneren in de assistentenkamer van Tergooi

Ziekenhuizen in Blaricum in 2011. Je eerste vraag was of ik onderzoek wilde doen. Hierna ben jij

van het eerst geschreven woord van de eerste beursaanvraag tot het laatst geschreven woord van

dit proefschrift zeer betrokken geweest. Onderzoek doen in het buitenland, ver van familie en

vrienden, was niet altijd even makkelijk en je bent me letterlijk overal gevolgd om steun, zowel

inhoudelijk als persoonlijk, te bieden waar nodig. Je was altijd bereikbaar, maar wist ook feilloos

wanneer ik ruimte nodig had. Ik neem jou (levens)lessen mee als het me in de toekomst lukt

om ook anderen te gaan begeleiden in het onderzoek. Ik hoop in ieder geval met je te kunnen

blijven samenwerken.

Mijn promotor, prof. dr. Grietje Molema. Beste Ingrid, dankzij Kate leerde ik je kennen in Boston,

alwaar je mijn poster stond te lezen in de gang van de CVBR. Ik had op dat moment geen promotor

en jij durfde het direct aan om die rol op je te nemen. Door jou zijn de verschillende hoofdstukken

in dit boekje tot een proefschrift geworden. Met jouw steun waagde ik me aan nog een avontuur

in Suriname. Voor jonge onderzoekers heb je de unieke gave om vertrouwen te geven terwijl

je ze blijft uitdagen en grenzen laat opzoeken. Hierin erken je ook altijd de persoon achter de

onderzoeker en geef je ze een eigen plekje. Gelukkig mocht deze onderzoeker bij jou en Nicoline

op de zolder slapen als ik weer eens in Nederland was met een doos met samples en zonder

dak boven mijn hoofd. En het was je dan zelfs niet teveel om in de ochtend boterhammen te

smeren om de dag op het lab mee door te komen. Ik hoop dat ik nog vaak langs mag komen voor

gezelligheid en wetenschap.

Mijn tweede copromotor, Dr. Matijs van Meurs. Beste Matijs, toen ik jou leerde kennen was het

heel fijn om de ervaringen uit Boston met je te kunnen delen en om te merken dat jij vergelijkbare

ervaringen hebt gehad. Vervolgens kon ik op jouw kennis en kunde bouwen om de uitdagende

projecten in Suriname tot een goed einde te brengen. Het was leuk om te merken dat je gaandeweg

steeds enthousiaster werd over de boodschap van mijn proefschrift. Hierdoor kreeg ik daar zelf

ook meer vertrouwen in. Uiteindelijk hebben gesprekken met jou een belangrijke rol gespeeld

in de keuzes die ik heb gemaakt voor de toekomst. Dank hiervoor. Ik hoop dat ik, naast voor

wetenschap, nog vaak langs mag komen voor je advies.

APPEN

DIX

III

177

&

Aan de leden van de beoordelingscommissie prof. dr. J Smit, prof. dr. J.G. Zijlstra en prof. dr. J. van

Woensel: hartelijk dank voor de vlotte en unanieme beoordeling van het proefschrift. I would also

like to express my gratitude to all co-authors who helped perform and improve research for this

thesis: prof. dr. Taco Kuijpers, dr. Nathan Shapiro, Nathanael Holband, Anna Bertoline, Francesca

Bardi, dr. Peter Dijk, Niek Achten, and Ellen Tromp.

Mijn paranimfen Philip Cotterell en Maurice Seleky, makkers van het eerste uur. Zonder onze

gezamenlijke geschiedenis was dit niet mogelijk geweest. Al bijna 30 jaar maken wij alles samen en

van dichtbij mee en deze dag hoort daarbij. Dank dat jullie mij willen bijstaan.

Rianne Jongman, lieve Rianne, dank voor jouw bereidheid om de uitdagende sample analyse met

mij uit te voeren voor dit proefschrift. Een analist is inderdaad de spil in een onderzoeksgroep. Je

hebt me heel veel vaardigheden in het laboratorium geleerd. Toen ik met jou aan de slag kreeg

ik weer plezier en vertrouwen in de pipet. Je was zelf bezig met je promotieonderzoek en toch

maakte je altijd tijd voor me als ik uit Suriname kwam met een doos met samples. Ik hoop dat

laatste nog vaak te kunnen doen en ook onze vriendschap te onderhouden.

In Suriname wil ik beginnen met het bedanken van Amadu Juliana. Lieve Amadu, dank voor het

creëren van een plek om voor het eerst dit soort onderzoek te doen in Suriname, en dank voor

jouw vriendschap. Met alles wat je hebt bereikt en doet blijf je uiterst bescheiden, en dat is zeer

leerzaam. Zoals jij mij begeleidde in de kliniek in Suriname, ben ik bereid om jou in je onderzoek

te begeleiden. Laten we elkaar vooral hier in Suriname tegen blijven komen om nieuwe projecten

te ondernemen, ook samen met Ming-Jan Tang. Lieve Ming, toen we elkaar in Suriname leerden

kennen waren we eigenlijk direct met elkaar bevriend. Ik denk dat het goed voor ons beiden goed

is geweest dat we elkaar hadden om te kunnen sparren over de projecten waar we mee bezig

waren. De uitdagingen waren enorm, maar het is ook jou gelukt om een stempel te drukken op de

zorg voor pasgeborenen in Suriname. Ik hoop in de toekomst veel met je te kunnen samenwerken

om deze lijn door te zetten. Hiervoor is de eerste stap gezet: Stichting P.R.E.S. Neirude Lissone

(boegbeeld van de NICU), Aartie Toekoen (baken van de pediatrie) en Shirley Heath, jullie ook

dank voor jullie deelname aan deze stichting en voor de prettige samenwerking en steun die ik van

jullie heb gehad in mijn tijd in Suriname. Het is altijd weer leuk om jullie te zien. Peter Moons, dank

voor de avonturen die we samen hebben beleefd in Paramaribo en de fietstochten daarbuiten.

Aan mijn collega arts-assistenten van de kinderafdeling Laurindo, Kevin, Safir, Charleen, Majenge,

Rhea, Louella, Natasha, Ritesh, Priya, Marit, Susanne, Femke, Pieter, en alle verpleegkundigen:

grantangi voor de samenwerking en het helpen met de inclusies voor de InSepSur studie. Jullie

hulp is onmisbaar geweest. Ik wil hier ook graag de ouders en pasgeborenen bedanken voor

deelname aan één van de studies.

Van het Scientific Research Center wil ik graag Wilco Zijlmans danken voor de ruimte die daar ik

kreeg om onderzoek te doen. Succes met de verdere opbouw van wetenschap in Suriname. We

APPEN

DIX

III

178

&

hebben samen drie mooie publicaties over Rhesus D van de grond gekregen met een significante

boodschap voor de zorg voor pasgeborenen in Suriname. Shellice, Anisma en Iris, volgens mij

ben ik de eerste die gaat promoveren, maar er zullen er nog vele volgen! Ik zal de ‘keek op

de week’ missen. Sigrid Ottevanger, ook onze banksessies onder de mangoboom zal ik missen,

maar ik hoorde dat we hetzelfde vak gaan uitoefen. Ik voorzie een vruchtbare toekomst!

Van het Academisch Ziekenhuis Paramaribo wil ik dr. John Codrington bedanken. Toen ik je leerde

kennen zette je direct de deur van het klinisch laboratorium wagenwijd open. Zonder jou en de

hulp van alle analisten in het laboratorium was het niet gelukt om deze resultaten te bereiken.

In het bijzonder wil ik ook Sheldon Simson, Judith Liong a Kong en Jimmy Roosblad hartelijk

bedanken voor onze directe samenwerking in het laboratorium. Met elkaar hebben we ook nog

een verfrissend project over het Zika virus tot twee prachtige publicaties gebracht. Dit laatste

was niet mogelijk geweest zonder dr. Stephen Vreden in Paramaribo en prof. dr. Jan Wilschut in

Groningen. Dank voor jullie steun in al mijn projecten en de samenwerking. Volgens mij zitten we

heel erg op één lijn en onze gesprekken hebben er mede toe geleid dat ik heb besloten om het

roer om te gooien en arts-microbioloog te worden. Voor dat laatste dank ik ook Sandra Hermelijn,

want ook jij zette de deur open en ik voelde me altijd zeer welkom op de koffie (met gebak) bij

jou op kantoor. Patricia Wong Lie Song en alle andere analisten van het serologisch laboratorium

van het AZP, dank voor jullie fantastische hulp met sample opslag voor de RheSuN studie. Voor dit

project wil ik ook graag alle verloskundigen en analisten van het AZP, RKZ, Diakonessen en ’s Lands

Hospitaal bedanken. Het was inspirerend om te zien hoe goed er werd samengewerkt en met wat

voor resultaat! Ik wil ook graag Margriet Lamers, Jedda Eppink, en Peter Schmitz bedanken voor

de hulp met dataverzameling. Ik had eigenlijk geen tijd, maar door jullie is het toch gelukt. Henk

Schonewille, Anneke Brand, en Humphrey Kanhai, ik denk dat we iets moois hebben neergezet

en dat we begonnen zijn om de zorg voor pasgeborenen in Suriname, op dit vlak te verbeteren.

Laten we vooral doorgaan. Ik dank het ministerie van Volksgezondheid van Suriname hierbij voor

de unieke kans om dit onderzoek te doen.

In the United States I would like to express my gratitude to the people from the Center for Vascular

Biology Research at Beth Israel Deaconess Medical Center in Boston. Dr. Christopher Carman

and dr. Roberta Martinelli, it all began in your lab. Thank you for letting me work there and learn

about the endothelium, neutrophils, microscopy, and science in general. Also, many thanks to

the members of the Carman lab Grace Teo and Peter Sage. Dr. Nathan Shapiro, thank you for

helping us with samples to analyse. Kate Spokes, thank you for guiding me those two years, our

coffee talks in the hallway of the CVBR, the dinner parties at your place, and for introducing me to

Ingrid. I am sure you and Ann and myself will stay in touch. To the members of the Aird and Dvorak

(in particular prof. dr. W.C. Aird and prof. dr. H. Dvorak), my next-door neighbours, thank you

for the two years of science and fun we had. Stanley, Salvatore and Brian, I had a great time with

you, both in and outside the lab. I still remember how you helped with an online application for

a job in the Netherlands. I did not get the job, but it doesn’t matter. I wish you all well in all your

future endeavours. I am also grateful for all of the friendships I made throughout my stay in Boston.

APPEN

DIX

III

179

&

Vincent, ik mis je, maar volgens mij gaat het heel goed met jou en Pauline en de kids. Ik zie je snel,

waar ook ter wereld.

Van Tergooi Ziekenhuizen Blaricum wil ik graag alle kinderartsen en arts-assistenten uit het

magische jaar 2011-2012 bedanken. Tevens wil ik Karen Verloop van het wetenschapsbureau

de stichting tot Bijstand Tergooi bedanken voor de inhoudelijke en financiële steun voor het

onderzoek en proefschrift. Van de kinderafdeling in het Reinier de Graaf Gasthuis in Delft wil ik

alle kinderartsen, in het bijzonder de opleider Boudewijn Bakker, bedanken voor de gelegenheid

om als arts-assistent te werken en de ruimte om mijn proefschrift af te ronden. Daarnaast

dank ik al mijn collega arts-assistenten voor de ruimte die jullie mij hiervoor hebben gegeven.

In het Universitair Medisch Centrum Groningen wil ik graag alle leden van de EBVDT groep

bedanken voor de vrolijke verwelkomingen en gastvrijheid tijdens de momenten dat ik op het

laboratorium was.

Aan al mijn dierbare vrienden en vriendinnen, ik ben even weg geweest. Iedere keer als ik even

terug was realiseerde ik me hoeveel ik jullie miste, maar merkte ik vooral dat de vriendschap weer

hechter was. Ik ben weer terug in Amsterdam. Laten we vooral doorgaan waar we gebleven waren.

Papa, mama, Erik en Janna. Dank voor de steun in de afgelopen vijf jaar. Jullie hebben me geleerd

om, ondanks tegenslag, toch vooral vooruit te blijven kijken en dat heeft geholpen. Opa, je blijft

mijn inspiratie, en nu op naar de 100 en verder. Lieve T, ik ken je al heel lang, maar sinds kort leer

ik je iedere dag weer beter kennen.

Rens Zonneveld

Paramaribo, oktober 2017

APPEN

DIX

IV

180

&

APPENDIX IV: LIST OF PUBLICATIONSVroon P, Roosblad J, Poeze F, Wilschut J, Codrington J, Vreden S, Zonneveld R. Severity of acute

Zika virus infection: an emergency room surveillance study during the 2015-2016 outbreak in

Suriname. Accepted for Publication.

Zonneveld R, Jongman RM, Juliana A, Molema G, Van Meurs M, Plötz FB. Early onset sepsis in

Surinamese newborns is not associated with elevated serum levels of endothelial cell adhesion

molecules and their shedding enzymes. Submitted.

Zonneveld R, Simson S, Codrington J, Juliana A, Plötz FB. Immature-to-total-granulocyte

ratio as a guide for antibiotic treatment in suspected early onset sepsis in Surinamese

newborns. Submitted.

Zonneveld R, Holband N, Bertolini A, Bardi F, Lissone N, Dijk P, Plötz FB, Juliana A. Improved

referral and survival of newborns after scaling up of intensive care in Suriname. BMC Pediatrics

2017 – Accepted for Publication.

Achten N, Zonneveld R, Tromp E, Plötz FB. Association between early onset sepsis calculator

and infection parameters for newborns with suspected early onset sepsis. Journal of Clinical

Neonatology 2017, 6:159-62.

Zonneveld R, Jongman RM, Juliana A, Zijlmans W, Plötz F, Molema G, Van Meurs M. Low serum

Angiopoietin-1, high Serum Angiopoietin-2, and high Ang-2/Ang-1 protein ratio are associated

with early onset sepsis in Surinamese newborns. Shock 2017, May 22.

Zonneveld R, Kanhai HHH, Lamers M, Brand A, Zijlmans CWR, Schonewille H. RhD

antibodies in pregnant women in multi-ethnic Suriname: The bservational RheSuN study.

Transfusion 2017, 57(10):2490-2495.

Zonneveld R, Lamers M, Schonewille H, Brand A, Kanhai HHH, Zijlmans CWR. Prevalence of positive

direct antiglobulin test and clinical outcomes in Surinamese newborns from RhD negative women.

Transfusion 2017, 57(10):2496-2501.

Zonneveld R, Schonewille H, Brand A, Kanhai, HHH, Zijlmans CWR. Evaluation of the presence of

clinically significant hemolytic disease of the fetus and newborn due to RhD antibodies in multi-

ethnic Suriname. Annals of Global Health 2016, 82(3):395-96.

Zonneveld R, Molema G, Plötz FB. Measurement of functional and morphodynamic neutrophil

phenotypes in systemic inflammation and sepsis. Critical Care 2016, 20:235-36.

APPEN

DIX

IV

181

&

Zonneveld R, Roosblad J, Van Staveren JW, Wilschut JC, Vreden SGS, Codrington J. Three atypical

lethal cases associated with acute Zika virus infection in Suriname. IDCases 2016, 5:49-53.

Zonneveld R, Schmitz P, Eppink J, MacDonald MS, Nahar - van Venrooij LMW, Kanhai HHH,

Zijlmans CWR. Rhesus D negativity amongst pregnant women in multiethnic Suriname.

Transfusion 2016, 56(2):321-4.

Zonneveld R, Molema G, Plötz FB. Analyzing neutrophil morphology, mechanics, and

motility in sepsis: options and challenges for novel bedside technologies. Critical Care

Medicine 2016, 44(1):218-28.

Zonneveld R, Martinelli R, Shapiro NI, Kuijpers TW, Plötz FB, Carman CV. Soluble adhesion

molecules as markers for sepsis and the potentia pathophysiological discrepancy between

neonates, children and adults, Critical Care 2014, 18(1):204-18.

Hew MN, Zonneveld R, Kümmerlin IP, Opondo D, de la Rosette JJ, Laguna MP. Age and gender

related differences in renal cell carcinoma in a European cohort. Journal of Urology 2012, 188(1):33-8.

APPEN

DIX

V

182

&

APPENDIX V: CURRICULUM VITAERens Zonneveld was born in Breda on the 8th of April 1983 and lived there for 18 years with his

parents and older brother. He completed the Gymnasium of Breda in 2001 before moving to

Amsterdam to study English Language and Literature in 2002. He started Medical School at

the Academic Medical Center (AMC) in 2004. During his studies, he was able to participate on

several research projects at the Departments of Vascular Medicine and Urology at the AMC. He

finished Medical School with an elective internship in Neonatology at St. Lucas Andreas Hospital

in Amsterdam. After he received his medical degree in December 2010, Rens started working

as a resident of pediatrics at Tergooi Hospitals in Blaricum. Here, he met dr. Frans Plötz who

encouraged him to continue research and supervised him on preparing his first research proposal

on human sepsis. He obtained funding from Tergooi Hospitals, ‘De Drie Lichten’ Foundation and

the Ter Meulen Fund, and received the Young Investigator Exchange Programme Award of the IPRF

European Society for Pediatric Research. This enabled him to start a research fellowship on the cell

biology of human sepsis in the laboratory of dr. Christopher Carman at the Center for Vascular

Biology Research at Beth Israel Deaconess Medical Center and Harvard Medical School in Boston,

USA. He received training in basic and specific laboratory skills, such as cell culturing, human

cell separation, flow cytometry, and fixed and live cell imaging. Here, he also met his promotor

prof. dr. Grietje Molema and copromotor dr. Matijs van Meurs. The knowledge he obtained on

cell biological and molecular aspects of leukocyte-endothelial interactions provided the rationale

behind the work on early onset sepsis (EOS) in newborns. He moved to Suriname in 2014 to start

working on clinical studies on EOS. Funding through the Thrasher Early Career Award enabled

setting up and executing the clinical studies on newborn EOS, in collaboration with the staff of

the Academic Hospital Paramaribo in Suriname and the University Medical Center Groningen. This

led to the various observational studies into the vascular pathophysiology of EOS, reported in

this thesis. He also participated as a physician in the transition of Suriname’s neonatal care facility

to a modern environment. He coordinated various other research projects in Suriname, such as

a nationwide evaluation of the presence of Rhesus D antibodies amongst pregnant women and

alloimmunization and hemolytic disease of the fetus and newborn in their offspring, and severity

of acute Zika virus infection during the recent outbreak. His main interest is to make clinically

feasible and affordable diagnostic tools for complex diseases, such as sepsis, become accessible

to developing countries. He intends to maintain his personal and professional relationship

with Suriname to continue to enhance care for its newborns and children. For this purpose he

co-founded the Pediatric Research & Education Suriname (P.R.E.S.) foundation together with

Surinamese and Dutch colleagues. He will start his residency to become a clinical microbiologist

at the Department of Microbiology of the AMC in January 2018. Rens loves travelling, fitness, road

cycling, and reading.


Recommended