+ All Categories
Home > Documents > RELATIONSHIP BETWEEN NUTRITIONAL STATUS AND THE …

RELATIONSHIP BETWEEN NUTRITIONAL STATUS AND THE …

Date post: 24-Nov-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
88
RELATIONSHIP BETWEEN NUTRITIONAL STATUS AND THE ABILITY OF SIX TO TWELVE- WEEKS INFANTS TO SERO-CONVERT TO ROTAVIRUS VACCINATION By Chishimba Katema A Dissertation submitted to the University of Zambia in partial fulfillment of the requirements of the degree of Masters of Medical Microbiology UNIVERSITY OF ZAMBIA Lusaka 2020
Transcript

RELATIONSHIP BETWEEN

NUTRITIONAL STATUS AND THE

ABILITY OF SIX TO TWELVE- WEEKS

INFANTS TO SERO-CONVERT TO

ROTAVIRUS VACCINATION

By

Chishimba Katema

A Dissertation submitted to the University of Zambia in partial

fulfillment of the requirements of the degree of Masters of Medical

Microbiology

UNIVERSITY OF ZAMBIA

Lusaka

2020

ii

DECLARATION

This Dissertation represents Chishimba Katema’s own work and has not been previously submitted

for a degree, diploma or other qualification at this or any other University.

Candidate Name: Chishimba Katema

Signature: ............................

Date: ............................

iii

CERTIFICATE OF APPROVAL

This dissertation of Chishimba Katema has been approved in partial fulfilment of the requirements

for the degree of Master of Medical Microbiology by the University of Zambia.

……………… ………………… …….………………

Supervisor Signature Month/Date/Year

……………… ………………… …….………………

Examiner 1 Signature Month/Date/Year

……………… ……………… …..…………………..

Examiner 2 Signature Month/Date/Year

……………… ……… ..…… …..………………..

Examiner 3 Signature Month/Date/Year

iv

DEDICATION

This dissertation is dedicated to my wonderful Family. I also dedicate this work to the Love of

My life, to all my children and my sisters.

v

ACKNOWLEDGEMENTS

Firstly, thanks and praises go to the Lord Almighty, Jesus Christ for He has been and always will

be faithful in all ups and downs. In no particular order, I would like to thank the following for

helping in my successful completion of this work. I would like to acknowledge the efforts and

contributions of my supervisors Dr Sody Munsaka, Dr Roma Chilengi and co-supervisor Dr.

Michelo Simuyandi who gave me valuable support regardless of their limited and precious time.

Other professional contributors to this work, Dr Samuel Bosomprah, Ms. Katayi Mwila

Kazimbaya, who offered their expert opinion in the design and implementation of this work. I am

grateful to the parents of all the infants who participated in this study.

vi

ABSTRACT

Live attenuated oral vaccines against rotavirus have been shown to be less efficacious in children

from developing countries compared to developed countries. Reasons for this disparity are not

fully understood.

This study aimed to investigate the potential association between indicators of nutritional status

(vitamin A status, weight for age, height for age and mid-upper arm circumference) of the infants

and sero-conversion among Zambian infants routinely immunized with rotavirus vaccine,

Rotarix™.

A total of 1320 infants were assessed for enrolment and420 infants were recruited at infant age

6-12 weeks in Lusaka, Zambia. Clinical information and samples were collected at baseline and

at one month following the second dose of rotavirus vaccine. Only 208 infant samples were

analyzed at baseline and one month after vaccination to determine infant nutritional status

(vitamin A status, weight for age, height for age status and mid-upper arm circumference.

Vitamin A status and serum rotavirus-specific IgA were determined using standardized Enzyme

Linked Immuno-sorbent Assay methods. The anthropometric indices were interpreted using

WHO growth standards. Sero-conversion was defined as a ≥ 4 fold rise in serum IgA titre from

baseline to one-month post Rotarix™ dose 2, while sero-positivity of IgA was defined as serum

titre ≥ 40. Pearson Chi-squared test was used to investigate the association of sero-conversion

and categorical factors (i.e. sex, vitamin A status, weight for age, height for age, mid-upper arm

circumference at baseline and serum IgA sero-positivity).

The sero-conversion frequency to rotavirus was 57.2% (119/208) and baseline infant sero-

positivity to rotavirus was 23.1% (48/208). Baseline vitamin A deficiency, underweight and

stunting were 78.9% (164/208), 6.7% (14/208) and 52.4% (109/208), respectively. Base-line

Infant moderate acute malnutrition was 38.0% (79/208208) and severe acute malnutrition was

12.50% (26/208) as determined by the infant mid-upper arm circumference. There was no

evidence of association between infant serum IgA sero-conversion and nutritional variables;

serum vitamin A deficiency (p=0.882), stunting (p=0.905), underweight (p=0.243), Mid-Uper

arm circumference (p=0.565) and infant baseline serum IgA sero-positivity (p=0.832).

Poor IgA sero-conversion frequency observed in this cohort was not influenced by nutritional

factors indicated by infant serum vitamin A status, weight for age, height for age and mid-upper

arm circumference. Early infant exposure to rotavirus infection, determined by infant serum IgA

sero-positivity levels did not influence sero-conversions frequencies in this cohort. These factors

may have other effects later in childhood, as no difference was observed among seroconvertors

and non-seroconvertors to rotavirus vaccination in the 6-12 weeks age group. Further research is

needed to better understand vaccine sero-conversion.

Key word: Rotavirus vaccine, rotavirus, seroconvert, Zambia, Infant

vii

TABLE OF CONTENTS

DECLARATION ................................................................................................................................... ii

CERTIFICATE OF APPROVAL ........................................................................................................ iii

DEDICATION ...................................................................................................................................... iv

ACKNOWLEDGEMENTS................................................................................................................... v

ABSTRACT .......................................................................................................................................... vi

TABLE OF CONTENTS .................................................................................................................... vii

LIST OF TABLES................................................................................................................................ ix

LIST OF FIGURES ............................................................................................................................... x

LIST OF ABBREVIATIONS............................................................................................................... xi

CHAPTER 1: INTRODUCTION ......................................................................................................... 1

1.1 Background ................................................................................................................................... 1

1.2 Statement of the Problem ............................................................................................................... 4

1.3 Justification of the Study ................................................................................................................ 5

1.4 Research Questions ........................................................................................................................ 5

1.5 Objectives ...................................................................................................................................... 6

1.5.1 General Objective ................................................................................................................... 6

1.5.2 Specific Objectives ................................................................................................................. 6

CHAPTER 2: LITERATURE REVIEW .............................................................................................. 7

2.1 Virology of Rotavirus .................................................................................................................... 7

2.1.1 Structure and Taxonomy ......................................................................................................... 7

2.1.2 Classification .......................................................................................................................... 8

2.1.3 Epidemiology ........................................................................................................................ 10

2.1.4 Replication Cycle .................................................................................................................. 11

2.1.5 Immunity to Rotavirus Infection ............................................................................................ 12

2.2 Rotavirus Acute Gastroenteritis (RV-AGE) .................................................................................. 14

2.3 Clinical Picture of Rotavirus Gastroenteritis ................................................................................. 15

2.4 Transmission and Shedding.......................................................................................................... 16

2.6 Rotavirus Vaccines ...................................................................................................................... 17

2.6.1 Rotashield™: Rhesus Rotavirus Vaccine ............................................................................... 18

2.6.2 Rotateq™: Bovine-Human Reassorted Rotavirus Vaccine ..................................................... 18

2.6.3 Rotarix™: Human Attenuated Rotavirus Vaccine .................................................................. 19

2.6.4 Rotavirus Vaccine Effectiveness ........................................................................................... 21

2.7 Nutrition Status ............................................................................................................................ 23

2.8 Weight for Age Status .................................................................................................................. 24

2.9 Height for Age Status................................................................................................................... 24

2.10 Mid-upper Arm Circumference (MUAC) ................................................................................... 25

viii

2.11 Serum Vitamin A Status of Children .......................................................................................... 26

2.12 Consequences of Under-nutrition on Mortality/Morbidity .......................................................... 26

2.13 Malnutrition and the Immune System ......................................................................................... 28

2.14 Malnutrition and Vaccination ..................................................................................................... 29

CHAPTER 3: METHODOLOGY ...................................................................................................... 31

3.1 Study Design ............................................................................................................................... 31

3.2 Study site ..................................................................................................................................... 31

3.3 Study Population.......................................................................................................................... 31

3.3.1 Inclusion Criteria .................................................................................................................. 32

3.3.2 Exclusion Criteria ................................................................................................................. 33

3.3.3 Sample Size Determination ................................................................................................... 33

3.3.2 Clinic Procedures .................................................................................................................. 34

3.4 Laboratory Procedures. ................................................................................................................ 35

3.4.1 Measurement of Serum IgA for the Determination of Sero-positivity and Sero-conversion .... 35

3.4.2 Measurement of Serum Vitamin A to Determine the Prevalence of Vitamin A Deficiency ..... 36

3.5 Data Analysis .............................................................................................................................. 36

3.6 Ethical Considerations ................................................................................................................. 36

3.7 Limitations .................................................................................................................................. 37

CHAPTER 4: RESULTS .................................................................................................................... 38

4.1 Patient Recruitment and Descriptives ........................................................................................... 38

4.2 Prevalence of Poor Nutritional Status in Infants and Infant Early Exposure to Rotavirus Infection 38

4.3 Effects of Nutritional Status on Sero-conversion Among Children 3 months in Lusaka Zambia .... 39

CHAPTER 5: DISCUSSION, CONCLUSION AND RECOMENDATIONS ................................... 42

5.1 Discussion ................................................................................................................................... 42

5.2 Conclusion................................................................................................................................... 45

5.3 Recommendations........................................................................................................................ 46

REFERENCES .................................................................................................................................... 47

APPENDICES ..................................................................................................................................... 61

Appendix A: Letters of support .......................................................................................................... 61

Appendix B: Study questionnaire and clinical examination form. ....................................................... 64

ix

LIST OF TABLES

Table 3.0 Z-score classification to determine nutritional status in children (WHO, 2006) ........... 34

Table 4.1 The prevalence of poor nutritional status in infants attending routine immunization at

Kamwala clinic ........................................................................................................................... 39

Table 4.2 Effects of nutritional status on sero-conversion among children 3 months in Lusaka Zambia .......................................................................................................................................... 41

x

LIST OF FIGURES

Figure 3: Recruitment and sample processing flow diagram .........................................................32

xi

LIST OF ABBREVIATIONS

AGE Acute gastroenteritis

DLPs Double layered particles

DRC Democratic Republic of Congo

DsRNA Double stranded ribonucleic acid

EED Environmental enteric dysfunction

ELISA Enzyme linked immune-sorbent assay

ER Endoplasmic reticulum

EPI Expanded Programme on Immunisations

FAO Food and Agriculture Organization

HAZ Height for age Z-scores,

HIC High income countries

HIV Human Imunodeficiency virus

IgA Immunoglobulin A

IgG Immunoglobulin G

IFN Interferon

IRF Interferon regulatory transcription factor

IIR Innate immune responses

LMIC Low and middle income countries

MHC Major histocompatibility complex

mL Millilitre

mRNA Messenger ribonucleic acid

NSP Non structural protein

xii

Nab Neutralizing anti bodies

OPV Oral polio vaccine

RV-AGE Rotavirus acute gastroenteritis

RNA Ribonucleic Acid

RVs Rotaviruses

SES Socioeconomic status

SG Sero-group

TB Tuberculosis

Th1 cells Helper T-cells

TGF-β Transforming growth factor beta

TLPs Triple layered particles

UNSCN United Nations Standing Committee on Nutrition

UNZABREC University of Zambia Biomedical Research and Ethics Committee

USA United States of America

VA Vitamin A

VAS Vitamin A supplementation

VE Vaccine efficacy

VP Viral protein

WAZ Weight for age Z-scores

WHO World Health organization

WHZ Weight for height Z-scores

μmol/L Micro-mole per litre

1

CHAPTER 1: INTRODUCTION

1.1 Background

Diarrhoeal diseases are one of the world’s leading killers of children and rotavirus is the most

common cause of severe diarrhoea among children under the age of five globally, and almost every

child is infected during the first five years of life (Tate et al., 2016). Rotavirus disease is typically

associated with vomiting and fever, followed by profuse watery diarrhoea, and the main cause of

death is dehydration. The most important therapy is oral or intravenous rehydration, as there is no

specific antimicrobial treatment for rotavirus disease. Although oral rehydration therapy had

reduced mortality by the time the first rotavirus vaccine was licensed in 2005, there still were about

450 000 deaths due to rotavirus gastroenteritis (RVGE) around the world (Tate et al., 2012). With

over 80% of those deaths occurring in low-income countries, rotavirus infection was so prevalent

that nearly every child in the world was estimated to have had at least one episode of rotavirus

disease by age two (Walker et al., 2013).

Globally, diarrhoeal diseases of any kind account for approximately 578,000 deaths among

children under the age of five. In this age group, rotavirus infection is still the leading cause of

severe acute gastroenteritis, accounting for an estimated 215,000 (37%) deaths annually and is

responsible for millions of hospitalizations and clinic visits (Tate et al., 2016, Walker et al., 2013).

Nearly 150,000 African children die from the dehydrating diarrhoea caused by rotavirus infection

every year, accounting for more than 56% of the global total of rotavirus deaths (WHO, 2013;

(Tate et al., 2016).

2

In Zambia, children under five years of age experience 10 million annual episodes of

diarrhoea, which result in an estimated 63,000 hospitalizations and 15,000 deaths (Liu et al.,

2015b, Mpabalwani et al., 1995). This makes diarrhoea Zambia’s third leading cause of under-5

mortality (after pneumonia and malaria) causing approximately 9% of deaths in children under

five years of age (Walker et al., 2013). One-quarter to one-third of the severe diarrhoea resulting

in hospitalization and death are attributable to vaccine-preventable rotavirus (Walker et al., 2013).

Thus, while recent years have seen significant reductions in morbidity and mortality due to

rotavirus infection, there is still much work to be done. Improvements in hygiene and sanitation

have been helpful in decreasing the incidence of many causes of diarrhoea; these measures alone

do not prevent rotavirus transmission (Armah and Binka, 2014). Rather, based on findings that

early rotavirus infections provide infants increased protection against subsequent bouts of disease

(Dennehy, 2008).The most important public health approach to fight the disease has been the

widespread use of immunizations. Researchers have tried various strategies since the 1980s to

develop effective vaccines (Cortese et al., 2013), and after two vaccines, Rotarix™

(GlaxoSmithKline, Biologicals) and RotaTeq™ (Merck & co, inc), were found to demonstrate

safety and efficacy in Europe and North America in the mid-2000s, efforts in the past decade have

focused on rolling them out them globally (Armah et al., 2016, Jiang et al., 2010). A third vaccine

called ROTAVAC™ was recently introduced. ROTAVAC™ is based on the116E rotavirus strain

and is manufactured by Bharat Biotech International Limited of India (Bhandari et al., 2014). In

efforts to battle the diarrhoeal scourge, in 2013, Zambia rolled out the Rotarix™

(GlaxoSmithKline, Biologicals) rotavirus vaccine in its Expanded Programme on Immunization

(EPI). Rotarix™ is an attenuated human oral vaccine based on the G1P[8] strain.

3

Its safety and efficacy have been established in healthy infants across multiple countries, including

Tanzania, Kenya and Guinea (Linhares et al., 2008, Ruiz-Palacios et al., 2006, Vesikari et al.,

2007). However, the vaccine has been found less efficacious among children in poor, developing

countries as compared with middle income and industrialized countries for reasons that are not yet

completely understood (Armah et al., 2010, Glass et al., 2006, Richardson et al., 2010). There is

substantial variation in vaccine effectiveness within developing world settings. A trial of Rotarix™

conducted in South Africa and Malawi demonstrated substantially better efficacy in South Africa

(76.9% efficacy) than in Malawi (49.4% efficacy) (Madhi et al., 2010). It has been proposed that

high background rates of severe rotavirus disease, other enteric co-infections, chronic diseases

such as Human Immunodeficiency Virus (HIV), Tuberculosis (TB) and recurrent malaria

prevalent in these populations, co-administration of rotavirus vaccine with Oral Polio Vaccine

(OPV), breastfeeding at the time of vaccination, and interference from passively acquired maternal

antibody, may all play a role in this reduced vaccine responsiveness (Jiang et al., 2010). Other

factors include; suspected high levels of environmental enteric dysfunction (EED), individual

genetic factors and malnutrition. We Hypothesized that poor infant nutritional status may affect

effectiveness of the rotavirus vaccine.

A child’s nutritional status is most commonly assessed through measurement of a child’s

weight and height, as well as through biochemical, clinical assessment. Underweight, stunting and

wasting are the most common clinical manifestations of malnutrition for which children are

screened in developing countries by anthropometry. Malnutrition is the most common form of

immunodeficiency and although the immunology of malnutrition remains poorly characterized

(Heilskov et al., 2014). The effects of malnutrition on vaccine efficacy, sero-conversion rates and

immunoglobulin titres have been the subject of extensive research for several decades and studies

show that the role of malnutrition on vaccine responsiveness still needs to be characterized (Savy

et al., 2009). Malnutrition also affects physical growth, morbidity, mortality, cognitive

4

development, reproduction, and physical work capacity (Prendergast, 2015). Malnutrition is

associated with impairments in mucosal barrier integrity, and innate and adaptive immune

dysfunction (Myatt et al., 2006). Malnutrition remains one of Zambia’s biggest public health

problems with about 40% of under five children being malnourished (Demographic, 2015). The

immunity that vaccines elicit is a selective pressure to which rotaviruses may adapt. Therefore the

different strains of the rotaviruses must be clearly identified to understand the different levels of

immunogenicity each or a group of strains have on the vaccines. Vaccine responsiveness is

measured through serum immunoglobulin titres, IgA and IgG titres are both used use to measure

the protectiveness of the vaccine. Different end points are chosen to depict sero-conversion rates;

usually the base-line and post vaccination time points are taken to measure the vaccine

immunogenicity (Jiang et al., 2010, Franco et al., 2006a, Clarke and Desselberger, 2014)

1.2 Statement of the Problem

Despite good progress with global introduction of the rotavirus vaccines (RV), diarrhoea is still a

leading cause of death in children under the age of 5 years, and as earlier mentioned, diarrhoea is

Zambia’s third leading cause of under-5 mortality (after pneumonia and malaria) causing

approximately nine percent of deaths in children under the age of 5 years (Walker et al., 2013).

Rotavirus takes the lives of over 3,600 Zambian children under the age of five each year and

accounts for approximately 40% of all under five diarrhoeal deaths and hospitalizations in Zambia

(Demographic, 2015). Following RV introduction, there has been a notable reduction in the

number of deaths due to diarrhoea (Tate et al., 2016). However, there is consistent evidence from

clinical trials that RV have lower efficacy in low and middle income countries (LMIC): efficacy

of vaccine is between 80-90% in high income countries (HICs) as compared to 40-60% in LMIC

(Cortese et al., 2013, Ruiz-Palacios et al., 2006, Vesikari et al., 2013). Many Hypotheses exist as

5

to why RV perform desperately poorer in LMIC as compared to HIC but none seem to evaluate

the role of poor infant nutritional status on RV1 effectiveness in LMIC like Zambia.

1.3 Justification of the Study

With the introduction of Rotarix™ (RV1) in its EPI, Zambian children under five years of age

have experienced a reduction in RV-AGE of 51% (Mpabalwani et al., 2016). Although the vaccine

has reduced under five RV-AGE, its effectiveness still remains low at only 57% (Beres et al.,

2016). Many factors have been proposed as to why Rotarix™ is not as effective as in other

countries but the potential role that indicators of nutritional status [vitamin A status, weight for

age (WAZ), height for age (HAZ) and mid-upper arm circumference of the infants (MUAC)] and

sero-conversion among Zambian infants routinely immunized with rotavirus vaccine, (RV1) have

not been explored. Knowing the exact causes of poor vaccine effectiveness would help further

reduce RV-AGE and help change policy in relation to food supplementation regimes before

vaccination.

1.4 Research Questions

1.4.1 What is the nutritional status (height for age, weight for age, mid-upper arm circumference

and serum vitamin A status) of infants receiving RV vaccines at the Kamwala Clinic in Lusaka,

Zambia?

1.4.2 Could the poor nutritional status of these infants be associated with low sero- conversion

rates after rotavirus vaccination?

6

1.5 Objectives

1.5.1 General Objective

To determine the relationship between poor nutritional status of infants and rotavirus vaccine

sero-conversion rates.

1.5.2 Specific Objectives

i. To determine the prevalence of early stunting, underweight, acute malnutrition determined

by MUAC and vitamin A deficiency in Zambian infants attending routine immunization at

Kamwala clinic by age three months.

ii. To determine any relationship between poor nutritional status and infant sero-conversion

following routine rotavirus immunization.

7

CHAPTER 2: LITERATURE REVIEW

2.1 Virology of Rotavirus

2.1.1 Structure and Taxonomy

RVs are non-enveloped double-stranded RNA (dsRNA) viruses that belong to the Reoviridae

family, with a characteristic morphology of wheel-like particles (as seen by electron microscopy)

from which their name is derived. The mature and infectious rotavirus particle is approximately

100 nm in diameter including the spikes, and composed of a three-layered icosahedral protein

capsid surrounding the genome (Greenberg and Estes, 2009). The viral genome is constituted by

11 segments of dsRNA encoding six structural proteins (VPs), which provide structural support

and mediate cell entry, and six non-structural proteins (NSPs), which are only produced by RVs

in infected cells and are implicated in viral replication, morphogenesis, and evasion of the host

immune response. Each segment encodes a single protein, except segment 11 that encodes two

proteins (NSP5 and NSP6, in some viral strains) (Greenberg and Estes, 2009). The infective virion

consists of three concentric protein layers that surround and cover its genome. The inner layer or

core is constituted by 120 copies of VP2 (scaffolding protein), and anchored to each segment of

dsRNA are VP1 (RNA-dependent RNA-polymerase) and VP3 (guanylyltransferase and

methylase), both proteins implicated in genome transcription. The intermediate layer surrounds

the core and it is composed of VP6 organized as pentamers and hexamers, giving rise to 132

channels of three classes that play an important role in the entrance of compounds to the capsid

and the export of newly formed mRNAs. VP6 is the most abundant structural protein and it is

assembled to form double layered particles (DLPs), which are non-infectious but transcriptionally

active (Angel et al., 2012). The outer capsid, part of the entire infectious triple layered particle

(TLP) is made up of two proteins, the calcium-binding glycoprotein VP7 and VP4 spikes, involved

8

in cellular attachment during infection, both of which induce neutralizing antibodies (Hu et al.,

2012).

The non-structural proteins NSP1-NSP6 are essential for rotavirus replication because they

modify the cell functions to enable the release of new virions from the infected cells. The NSP1

protein is engaged in inhibition of IFN-α responses, NSP2 is required for dsRNA synthesis, and

NSP3 is essential for translational regulation and inhibition of host protein synthesis. The viral

enterotoxin NSP4 increases the concentration of Ca2+, which disrupts the cytoskeleton of

microvilli and the cellular homeostasis of the host. NSP4 is the major contributing factor to

electrolyte and fluid malabsorption causing diarrhea. In addition, NSP5 interacts with NSP2 to

form cytoplasmic structures known as viroplasms, inside of which RNA replication and

morphogenesis of new viral particles take place. NSP6 interacts with NSP5 in the viroplasms, but

its function is unknown, being not coded by all rotavirus strains (Hu et al., 2012).

2.1.2 Classification

RV strains have a high genomic and antigenic diversity that can be classified into four

different specificities: group, subgroup, serotype and genotypes (Matthijnssens et al., 2012). The

VP6 protein, the major capsid viral protein, confers the group specificity; groups (A-H) have been

established according to antigenic properties and sero-epidemiological studies (Greenberg and

Estes, 2009, Matthijnssens et al., 2012). However, only groups A-C and H can infect humans,

group A viruses being responsible for over 90% of all infections and this group is considered a

relevant target for vaccination (Greenberg and Estes, 2009), based on the amino acid sequences of

their VP6 protein, RVs can be grouped into subgroups (SG) and can be referred to as SG-I, SG-II,

SG-I/II, and non SGI/ II, depending on the presence or absence of subgroup-specific epitopes.

(Matthijnssens et al., 2012).

9

Within each group, rotaviruses can be further classified into serotypes defined by reactivity

of viruses in plaque reduction neutralization assays using polyclonal or monoclonal antisera raised

against the viral capsid proteins VP7 and VP4 (Marcelin et al., 2011). Given that the genes

encoding these proteins can segregate independently, a dual nomenclature system was established

in which the serotypes determined by the VP7 protein (termed G serotypes because VP7 is a

glycoprotein) and the VP4 serotypes (designated P serotypes since VP4 is protease-sensitive) are

considered (Matthijnssens et al., 2008). The G- and P-genotyping system is based on reverse

transcription polymerase chain reaction (RT-PCR), where different genotypes may be recognized

by their length and further sequenced (Matthijnssens et al., 2011b). VP7 types are classified as

serotypes by neutralization assays or as genotypes by sequencing; these 2 assays yield concordant

results, so viruses are referred to by their G serotype alone (e.g., G1, G2, G3, and so forth). VP4

serotypes are also classified by neutralization and sequencing assays, but the results do not always

agree, so there is a dual system for P typing. P serotypes are referred to by their serotype numbers

(e.g., P1, P2, and so forth) and P genotypes are denoted in brackets (e.g., P[8], P[4], and so forth).

G genotyping is the most widely used method for classification because of difficulties in

standardizing VP4 serotype assays. Currently, 27 G types (according to the nucleotide sequence

of VP7) and 37 P types (according to the nucleotide sequence of VP4) are known (Matthijnssens

et al., 2011a)

Rotaviruses may also be classified by their whole genome, where genome segments forVP7-

VP4-VP6-VP1-VP2-VP3-NSP1-NSP2-NSP3-NSP4-NSP5/6 are represented by the acronym Gx-

P[x]-Ix-Rx-Cx-Mx-Ax-Nx-Tx-Ex-Hx (where x= an Arabic numeral≤1) (Matthijnssens et al.,

2014). Each of the nine other internal gene segments (other than G- and P-typing gene segments)

have more than 8 genotype alternatives. Sequencing of the full genome has revealed that the

internal gene segments of the most common genotypes with P[8] P-type (G1P[8], G3P[8], G4P[8],

10

and G9P[8]) usually belong to genogroup 1, whereas the internal gene segments from G2P[4]

strains belong to genogroup 2 (Maes et al., 2009, McDonald and Patton, 2008). In addition,

phylogenetic analyses have revealed that the human genogroup 1 rotaviruses have developed from

the same origin as porcine rotaviruses, whereas the genogroup 2 viruses have a link to rotavirus

strains of bovine origin (Matthijnssens et al., 2014).The most common G and P type associations

worldwide related with RV infection in humans are G1P1A[8], G2P1B[4], G3P1A[8], G4P1A[8]

and G9P1A[8] (Matthijnssens et al., 2008, Matthijnssens et al., 2011a).

Sequencing of the full genome has revealed that the internal gene segments of the most

common genotypes with P[8] P-type (G1P[8], G3P[8], G4P[8], and G9P[8]) usually belong to

genogroup 1, whereas the internal gene segments from G2P[4] strains belong to genogroup 2

(Matthijnssens et al., 2014). In addition, phylogenetic analyses have revealed that the human

genogroup 1 rotaviruses have developed from the same origin as porcine rotaviruses, whereas the

genogroup 2 viruses have a link to rotavirus strains of bovine origin (Matthijnssens et al., 2014).

2.1.3 Epidemiology

Before rotavirus vaccinations in 2000-2004, rotavirus infections were estimated to cause over

500,000 deaths annually worldwide (Parashar et al., 2006). The majority of rotavirus related deaths

occur in developing countries (especially in India and Africa), where access to health care is

limited (Desselberger, 2014). Each child is normally infected at least once before the age of five,

the majority of them before two-years of age. Neonatal and adult RV infections are more

uncommon and often asymptomatic. In regions with a temperate climate, such as Europe, RV has

a clear seasonal distribution, the most active months being in the winter and/or early spring. In

subtropical and tropical climates the distribution of RV disease is not as clear as in Europe; the

most active months are during the cool and dry season, but sporadic infections may be detected

11

during the whole year (Chilengi et al., 2016, Hashizume et al., 2008, Luchs et al., 2014, Parashar

et al., 2006)

2.1.4 Replication Cycle

After ingestion, rotaviruses infect the mature enterocytes at the tip of the villi of the small

intestine and replicate in the cell cytoplasm. Upon contact with the cellular receptor, the VP4

spikes of RV three layered particles (TLPs) undergo conformational changes in such a way that

the lipophilic domains of VP5* which are normally hidden below VP8* are exposed on the surface

in form of a ‘post-penetration umbrella’ conformation (Kim et al., 2010, Settembre et al., 2011,

Trask et al., 2010). Following binding, the mechanism of cell penetration of RV particles remains

unclear; it may occur by receptor-mediated endocytosis or direct membrane penetration, with

solubilization of the outer capsid proteins VP7 due to low Ca2+

concentrations in endosomes to

yield double layered particles (DLPs) in the cytosol (Wolf et al., 2011). In the transcriptionally

active DLPs, the dsRNA is used as template and the VP1 protein produces positive sense, capped,

non-polyadenylated transcripts (McDonald and Patton, 2011). These transcripts are used as

templates for translation and for strand synthesis in the replication process to generate a complete

set of eleven dsRNA genome segments. RNA molecules are ejected from DLPs into the cytoplasm

through a series of channels (Desselberger, 2014, Trask et al., 2010). After translation, the

structural proteins VP1, VP2, VP3, and VP6; the non-structural proteins NSP2 and NSP5, and

RNA molecules aggregate in viral inclusion bodies (viroplasms) located in the cytoplasm. In these

viroplasms take place replication, RNA packaging, and DLPs assembly. Viral proteins VP4, VP7,

NSP1, NSP3, and NSP4 are not present in the viroplasm but are necessary for other processes.

NSP4 is attached to the endoplasmic reticulum (ER) membrane and binds VP6 in DLPs to facilitate

its translocation to the intra luminal side of the ER. There, DLPs are covered with VP7 and VP4

12

(by an incompletely understood mechanism) to give rise to TLPs, which will be expulsed from the

ER and subsequently from the cell before or during cell lysis (Criglar et al., 2014).

2.1.5 Immunity to Rotavirus Infection

Upon RV infection, acquired immune responses are elicited, both from B cells producing

antibodies directed against virus-specific proteins, and from T cells recognizing T cell-specific RV

epitopes on the surface of infected cells in complexes with major histocompatibility complex

(MHC) classes I and II antigens. Many antibodies directed against VP7 andVP4 on the surface of

RV particles are neutralizing antibodies (NAb) in vitro and protective in vivo, as demonstrated by

passive transfer in mice and gnotobiotic piglets as animal models (Yuan et al., 2008).

Passive transfer of RV-specific cluster of differentiation T cells (CD8+

) has also shown to be

protective. Transplacentally acquired RV-specific antibodies likely protect new-borns from

infection (Ray et al., 2007) and interfere with immune responses to RV vaccination (Appaiahgari

et al., 2014, Chilengi et al., 2016, Johansson et al., 2008). The availability of various knockout

mutant mice has permitted researchers to dissect the relative contributions of humoral and cellular

immune responses to protection while RV-specific T cells help eliminate RV after primary

infection, it is the RV-primed memory B cells which provide more long-term protection (Franco

et al., 2006b). Humoral antibodies boosted after repeated infection are directed against both

serotype-specific and cross-reactive epitopes on VP4 and VP7 molecules, thus also providing

heterotypic protection (Franco et al., 2006b). Plasma-cytoid dendritic cells were found to be

necessary and sufficient to induce B cell activation after RV infection in mice (in vivo) and

inhuman cells (in vitro) (Deal et al., 2013). Human RV-specific CD4+

T-cells circulating in the

blood express the intestinal homing receptor α4β7 (Parra et al., 2014). However, protection from

RV infection is not entirely correlated with the concentration of VP4- and VP7-specific NAbs.

Upon natural infection by or vaccination with RV, infants and young children develop antibodies

13

against other structural (VP6, VP2) and non-structural (NSP4) proteins. VP6-specific antibodies

do not neutralize in vitro, but were shown to be protective in vivo (Weitkamp et al., 2005), when

VP6-specific antibodies of the IgA class were applied. It was suggested and has recently been

proven that VP6-specific IgA antibodies are taken up (‘transcytosed’) by epithelial gut cells

through J protein receptors at the basolateral membrane and form complexes with new DLPs

released from viroplasm, thus preventing their maturation to TLPs (‘intracellular neutralization’)

(Aiyegbo et al., 2013, Sapparapu et al., 2014). However, it has also been shown that mice and

patients who are IgA-deficient eliminate RV after infection, probably due to a compensatory IgG

protective immunity (Corthesy et al., 2006). Passive transfer of NSP4 antibody has produced some

protection in mice (Hou et al., 2008), but actively acquired NSP4 antibody does not protect

gnotobiotic piglets from a challenge with RV (Aiyegbo et al., 2013). Prospective studies have

shown that, after 1 or 2 natural RV infections, children appear to be highly protected against severe

disease following infection by various, also heterotypic, RVs (Verkerke et al., 2016)

RV infection immediately triggers various mechanisms of innate immune responses (IIR),

which occur earlier than acquired RV-specific immune responses, at least after primary infection

(Angel et al., 2012). Many immune-inflammatory responses (IIRs) appear to be RV strain-specific

and also cell type-specific (Feng et al., 2009). At present it is not fully clear to what extent the IIRs

after RV infection modify disease outcome. It has been shown that the RV NSP1 is able to interact

with the following cellular proteins: interferon (IFN) interferon regulatory transcription factor

(IRF) 3 (Sen et al., 2011). NSP1 targets the pro-apoptotic cellular protein p53, leading to its

proteasomal degradation and thus delaying cell death during the early stages of RV replication

(Bhowmick et al., 2013).

14

In studies using animal models, rotavirus infection has been shown to induce type I (IFN-γ)

and type III (IFN- λ) interferon (IFN) messenger ribonucleic acid (mRNA) expression, which

reduces viral replication (Pott et al., 2011). In addition, the capacity of RV strains to inhibit the

IFN system has been shown to affect the degree of extra-intestinal spread of the virus in mouse

models. Although rotaviruses may potentially inhibit all types of IFN response by inhibiting their

transcription factors (IRF3, IRF5, IRF7, and NF-κВ) by NSP1 or inhibiting their signal complexes

(STAT1 and STAT2) (Sen et al., 2011), the human RVs have been shown to inhibit the IFN

response less efficiently than the animal RV strains (Rodriguez-Limas et al., 2014). In addition,

rotaviruses potentially inhibit the capacity of dendritic cells to activate type1 helper T-cells (Th1

cells) by stimulating secretion of a regulatory cytokine, transforming growth factor beta (TGF-β),

in Caco-2 cells in vitro (Feng et al., 2009).

2.2 Rotavirus Acute Gastroenteritis (RV-AGE)

Acute gastroenteritis (AGE) is one of the infectious disease entities that cause major morbidity

and mortality in the world. After pneumonia and malaria, diarrheal diseases are the third most

important infectious cause of death in children under the age of 5 (Liu et al., 2015a, Walker et al.,

2013). It is estimated that AGE causes 580 000 – 750 000 deaths in children of this age in the

world every year (Liu et al., 2015a). Most of these fatal cases occur in countries with poor health

care systems and a lack of safe water, sanitation and poor hygiene e.g. in Sub-Saharan Africa and

Southeast Asia. In developed countries, deaths due to AGE are very rare. However, AGE viruses

circulate in both developed and poor countries, causing an estimated 1.5 billion cases every year

in children and adults (Mandeville et al., 2009). As a consequence, the disease burden, morbidity,

heath care utilization and expenses caused by AGE is enormous.

15

Rotavirus acute gastroenteritis (RV-AGE) is a serious global issue that is associated with

substantial morbidity and mortality among infants and young children. In 2013, an estimated

47,100 rotavirus deaths occurred in India, which represented 22% of all deaths due to rotavirus

that occurred globally that year. Four countries (India, Nigeria, Pakistan, and Democratic Republic

of Congo) accounted for approximately half (49%) of all rotavirus deaths in 2013, and 10 countries

(India, Nigeria, Pakistan, Democratic Republic of Congo, Angola, Ethiopia, Afghanistan, Chad,

Niger, and Kenya) accounted for almost two-thirds of all deaths (65%) in 2013 (Tate et al., 2016).

Tate and co-workers estimated that 37% of the 578 000 diarrheal deaths in children <5 years of

age in 2013 were due to rotavirus, resulting in 215 000 rotavirus deaths in this age group (Tate et

al., 2016).

2.3 Clinical Picture of Rotavirus Gastroenteritis

The symptoms of RV-AGE consist of watery diarrhoea, vomiting and fever. Because of the

wide spectrum of symptom severity, the causative agent of an individual AGE episode cannot be

reliably differentiated by its symptoms (Gray et al., 2008). In the bigger picture, RV-AGE is often

more severe compared to AGE caused by other viruses: the risk of dehydration is higher, fever is

seen more often, and RV-AGE more often leads to hospitalization (Aupiais et al., 2009, Giaquinto

et al., 2007). Clinically, RV-AGE caused by different genotypes cannot be distinguished, although

it has been proposed that G1P[8] and G9P[8] cause more severe symptoms on average than other

RV genotypes. Diarrhoea with or without vomiting is the predominant feature in RV-AGE. In a

Finnish community-based prospective study of AGE in children less than two years of age, it was

showed that there was a difference between AGEs caused by different viruses: diarrhoea was more

pronounced in RV-AGE, whereas vomiting was seen more often in norovirus AGE (Greenberg

and Estes, 2009). The incubation period of RV-AGE is typically one to three days, with an average

of two days (Aupiais et al., 2009, Desselberger, 2014, Lee et al., 2013). The symptoms begin

16

acutely, most often with vomiting followed by watery diarrhoea. Sometimes stomach ache

precedes the major symptoms. Vomiting lasts on average for two to three days, and diarrhoea for

four to five days (Desselberger, 2014), but there is a great deal of variation. Fever is often seen,

and febrile seizures are seen in children with a predisposition for them. Bloody diarrhoea is rare.

Antigenaemia and viraemia can be seen but systemic complications are rare (Blutt et al., 2007).

Many children, however, are infected by RV without marked clinical symptoms. Especially infants

less than six months of age seem to have protection by maternal antibodies, and if infected during

these first months, the infection often remains silent (Desselberger, 2014). Also in older children

RV infection can sometimes be passed with nonspecific symptoms or no clinical symptoms at all

(Desselberger, 2014), usually if the child has already had a primary RV infection (Franco et al.,

2006b).

2.4 Transmission and Shedding

The main route of transmission of RV-AGE is faecal-oral route (Dormitzer et al., 2004), but

it can also occur via respiratory droplets containing mucus and viruses (Aupiais et al., 2009). The

spreading of the infection is very difficult to control. The virus is shed in large numbers, up to 1011

infectious viral particles/mL in stools of an infected person and, on the other hand, the contagious

dose of RV is very small: about 10 viral particles (Aupiais et al., 2009). Besides these, knowing

that RVs are highly resistant to environmental factors such as temperature and pH, it is easy to

understand why RV-AGE is very easily transmitted, especially at day-care centres, where young

children prone to infection are in close contact with each other, or in a hospital ward treating

children with and without AGE. Shedding of viruses in stools has been demonstrated for about

two or three weeks by reverse transcriptase polymerase chain reaction (RT-PCR), but in immune-

compromised persons the shedding can last as long as three months (Corcoran et al., 2014,

Mukhopadhya et al., 2013).

17

2.5 Diagnosis of Rotavirus Gastroenteritis

The presence of RV is commonly detected in stools by the Enzyme Linked Immuno-sorbent

Assay (ELISA) antigen test or RT-PCR. ELISA tests are commercially available and fast to

perform. The sensitivity of ELISA tests is good (Greenberg, 2011), but false positives can also

occur. Compared to the ELISA test, RT-PCR detects more RV-positive in stool specimens, the

sensitivity of the two methods is about the same in detecting severe cases. This suggests that the

sensitivity of ELISA is good enough to be used at the hospital level and in vaccine efficacy studies,

and the studies conducted using ELISA are comparable to the ones using RT-PCR (Desselberger,

2014). However, the ELISA cannot be used if the size of the stool sample is very small or if the

stool is soaked into a diaper. Furthermore, ELISA cannot determine RV genotype. Compared to

ELISA, RT-PCR is better in detecting RVs of genotype G2 and G4, and perhaps of more

uncommon strains as well. The RT-PCR method is highly sensitive, and the test can be done from

a minimal amount of stool, such as from diapers or a rectal swab. The genotype of RV can be

determined by sequencing the VP7 and VP4 coding regions. Until lately, RT-PCR testing has

mainly been used in research, but along with rapid-PCR techniques, the test is becoming more

commonly available, also in clinical practice (Desselberger, 2014).

2.6 Rotavirus Vaccines

The RV vaccines in clinical use are orally administered live vaccines with re-assorted human-

animal RVs or attenuated human RVs. The efficacy of these vaccines relies upon heterotypic cross-

immunity: after a natural or introduced infection caused by one RV type, the induced immunity

protects against consecutive RV infections, and after two infections the protective immunity

against moderate to severe RV AGE – also against heterotypic RVs – is excellent and long-lasting

(Fischer et al., 2002). Of the five most common RV genotypes in Western countries (G1P[8],

G9P[8], G2P[4], G3P[8] and G4P[8]), the G2P[4] genotype has been suspected to have the ability

18

to escape vaccine-induced cross-immunity better than the other common RV genotypes (Franco

et al., 2006b, Ruiz-Palacios et al., 2006). However, RV vaccines still show efficacy against severe

AGE associated with G2P[4] (Vesikari et al., 2007).

2.6.1 Rotashield™: Rhesus Rotavirus Vaccine

The first vaccine against RV that became commercially available was derived from rhesus

monkey RV G3P[3] re-assorted with human VP7 antigens G1, G2 and G3. The full vaccine series

consisted of three doses of oral live vaccine scheduled at two, four and six months of age. The

vaccine was highly reactogenic and induced febrile reactions in many vaccines. This vaccine,

tetravalent rhesus RV (RRV-TV) or Rotashield™, was launched in the USA in 1998. It was

recommended in the routine immunization of children in the above-mentioned schedule, and a

catch-up programme of children up to nine months of age in the first year. About 600 000 children

did enter the immunization program until the vaccine was withdrawn in 1999 due to association

with vaccination and intestinal intussusception. After withdrawal, in further studies, the relative

risk of intussusception following vaccination was assessed to be 58.9% (Rosillon et al., 2015) or

between 1/10 000 and 1/32 000 vaccinated children. The risk was associated with the age of the

vaccinated child: if the first dose of vaccine was given at between three and nine months of age,

as in the catch-up schedule, the risk of intussusception was elevated (Lynch et al., 2006, Simonsen

et al., 2005). The observed efficacy of Rotashield™ against severe rotavirus AGE was 82- 91%

against severe AGE, and efficacy against any RV AGE was 57-66% (Vesikari et al., 2006).

2.6.2 Rotateq™: Bovine-Human Reassorted Rotavirus Vaccine

Shadowed by Rotashield™, the second generation of RV vaccines were exposed to extensive

preclinical trials before entering clinical practice. Rotateq™ is a five-component human-bovine

re-assortant rotavirus vaccine in which human RV VP7 capsid proteins G1, G2, G3 and G4 are re-

assorted with bovine virus expressing P7[5], and one human VP4 P-type P[8] re-assorted with

bovine VP7 G6 (Hemming and Vesikari, 2014, Vesikari et al., 2006). The result is “pentavalent”

19

(RV5) vaccine. The full vaccination series consists of three doses. The first dose is given between

six and twelve weeks of age, and the subsequent doses at 4-10-week intervals until the third dose

is administered before 32 weeks of age. Diarrhoea, vomiting and low-grade fever, associated with

vaccination with RV5 have been reported at a similar or slightly higher rate in vaccinated and in

placebo recipients (Block et al., 2007). More severe diarrhoea associated with the emergence of a

human-bovine double re-assortant RV in vaccinated has been seen in case reports (Hemming and

Vesikari, 2014, Markkula et al., 2014). The major adverse effect of interest, intussusception, has

been continuously studied. A small risk has been found of intussusception associated with 3-7 days

after the first dose of RV vaccine. The relative risk of intussusception associated with RV5 has

been between 2.9 and 9.9 after the first dose, accounting for 0.79-7.0 excess cases of

intussusception per 100 000 children (Haber et al., 2008).

2.6.3 Rotarix™: Human Attenuated Rotavirus Vaccine

The other currently available vaccine against RV is also an orally administered live vaccine.

Rotarix™ has been attenuated from human RV-type G1P[8], and it is a “monovalent” vaccine

(RV1). The vaccination consists of two doses given between 6 and 24 weeks of age with at least

four-week intervals between doses. Adverse effects associated with RV1 have been sparse.

Irritability and vomiting have been seen, but no significant association with fever reactions

(Vesikari et al., 2013). Recently, a similar slightly elevated risk of intussusception following the

first dose of RV1 vaccine than with RV5 has been noted, the relative risk being between 1.1 and

6.8 (Rha et al., 2014), accounting for 1.2 to 2.8 per 100 000 children after the first dose of RV1

(Haber et al., 2015). The efficacy of RV1 against severe rotavirus AGE has been about 90% and

against all AGE between 58% and 90%, with the lowest efficacy against G2P[4]. (Parashar et al.,

2006, Ruiz-Palacios et al., 2006).

20

In India, candidate rotavirus vaccines are being developed using two strains (116E strain and

I321 strain) isolated from new-borns. The 116E strain is a P8[121] G9 natural re-assortant between

a human parent strain and a VP4 gene of bovine origin. This strain was isolated in 1985 from an

outbreak of asymptomatic rotavirus infections in New Delhi (Bhandari et al., 2014). The sequence

of the VP4 gene is homologous to that of P[11], a genotype commonly found in cattle. In addition,

the I321 strain was identified from an outbreak of a nosocomial infection at a maternity centre in

Bangalore and was identified to be a bovine-human re-assortant strain (Bhandari et al., 2014). The

genome of the I321 strain is different from the genome of the 116E strain. The I321 strain includes

nine bovine gene segments. Among them, only gene segments five and seven, which encoded non-

structural proteins 1 and 3, are of human origin. A new strain with the identical G and P segments

as the I321 strain has emerged in Vellore, India as a cause of gastroenteritis in children (Bhandari

et al., 2014).

Published results from a phase 3 clinical trial of an oral, attenuated rotavirus vaccine

(ROTAVAC™, Bharat Biotech International, Limited, of India), manufactured in India and based

on the natural human-bovine reassortant strain G10P (Kawamura et al., 2011), which causes

asymptomatic infection in neonates, demonstrated that the vaccine effectiveness for protection

from severe rotavirus gastroenteritis in the first year of children’s lives was 56% (Bhandari et al.,

2014). The results from the Indian multicentre trial showed that this vaccine efficacy was

comparable to that of the 2 internationally licensed rotavirus vaccines in low-income settings, and

alternate dosing schedules, including neonatal dosing schedules, are being evaluated as well

(Armah and Binka, 2014, Leshem et al., 2014). According to this observation, both commercially

available vaccines have been shown to be highly effective against severe rotavirus disease, despite

one being monovalent and the other pentavalent (Payne et al., 2013). This is important because

data from countries in Asia and Africa show greater strain diversity with several rotavirus types

circulating simultaneously (Todd et al., 2010).

21

2.6.4 Rotavirus Vaccine Effectiveness

The World Health Organization (WHO) in 2009 issued a global recommendation for the

inclusion of rotavirus vaccines into routine immunization programs, based on results from

successful clinical trials from Asia and Africa. (Agócs et al., 2014, Danchin and Bines, 2009).

These vaccines have great potential to prevent the severe morbidity and mortality from rotavirus,

but studies consistently demonstrate a gradient of reduced efficacy in low socio-economic settings

(SES) where the burden of severe rotavirus disease, particularly mortality, is greatest (Nelson and

Glass, 2010). Clinical trials and observational studies of oral rotavirus vaccines performed in

infants in high income settings demonstrated vaccine efficacy (VE) exceeding 90% (Boom et al.,

2010, Buttery et al., 2011). In middle income settings of Latin America, South Africa and Vietnam,

VE ranged from 72 to 83%, (Madhi et al., 2010) while in low income settings in Asia and Africa,

VE ranged from 39 to 49% and 57% in Zambia (Beres et al., 2016). A research after the

introduction of RV1 in Zambia’s EPI recorded declines in RV-AGE in infants (Mpabalwani et al.,

2016). In the same study it was observed that a marked reduction in RV-AGE occurred in the

rotavirus positivity rate from 44.6% to 26.2% in the pre– and post– rotavirus vaccine eras,

respectively, representing a reduction of 51%. These same patterns are apparent for both currently

licensed vaccines (single-strain Rotarix™ (GlaxoSmithKline Biologicals) and pentavalent

Rotateq™ (Merck & Co) referred to as RV1 and RV5, respectively). Understanding the biological

basis for this poorer performance may be crucial for maximizing the impact of current vaccines

and for guiding the development of new ones.

With all existing live oral vaccines against enteric infections (including typhoid, cholera and

oral polio), the immune response and efficacy are diminished amongst certain populations living

in developing countries (Pasetti et al., 2011). While the exact reasons for this phenomenon are

unclear, a range of hypotheses has been proposed. These can be broadly categorized as (1) factors

22

leading to a poor immune response to natural infection, (2) reduced immunogenicity of the

vaccines, and (3) very high incidence rate of infection that overwhelms immunity from vaccination

(Moon et al., 2010). In very young infants in low Social and Economic Status (SES), greater levels

of maternal antibody acquired trans-placentally or from breast milk may serve to neutralize vaccine

virus so as to reduce replication and antigen load and thereby the decrease the Immunogenicity

(Chilengi et al., 2016). The greater diversity of rotavirus strains circulating in many developing

countries may also lead to weaker natural and vaccine-derived immunity (Santos and Hoshino,

2005). Furthermore, micronutrient deficiency and nutritional status are specific factors that have

all been associated with diminished immune response to live oral vaccination, and would also

affect immunity to natural infection (Ahmed et al., 2009). The extent to which nutritional factors

affects vaccine responsiveness still remains poorly characterized and identifying whether

nutritional factors can explain the reduced vaccine efficacy observed in low socioeconomic status

(SES), and which factors are most important, can help to prioritize future research aimed at

improving vaccine performance.

2.6.4.1 Rotavirus Vaccines in Zambia

The Zambian government, in 2012 initiated a 2-year pilot introduction of the RV1, oral

rotavirus vaccine in all public health facilities in Lusaka Province and in 2013 introduced it in the

routine immunization programme (Chilengi et al., 2016). The RV1 is given at 6 and 10 weeks of

age (without a catch-up dose). The first dose of monovalent rotavirus vaccine is administered early

in life (6 weeks) due to early exposure of rotavirus infection and allowing the first dose to be

administered between 6 and 20 weeks of age. (Chilengi et al., 2016). Following the introduction

of RV1 in the national EPI, a research was conducted to estimate the vaccine effectiveness and the

overall trend of VE, ranged from 17% to 60% for at least 1 dose, this was consistent with other

studies from other developing countries (Mpabalwani et al., 2016). These findings support prior

23

reports of the relatively lower VE of live oral vaccines in low- to middle-income countries such as

Zambia, compared with more developed countries (Madhi et al., 2010).

Another study conducted the University Teaching Hospital (UTH) after the introduction of

RV1 in Zambia’s EPI recorded declines in, all-cause diarrhoea and rotavirus AGE hospitalizations

as well as in-hospital diarrhoea deaths as alluded to earlier (Mpabalwani et al., 2016).

2.7 Nutrition Status

Nutritional status of a population is a prominent reflection of a nation’s economic

development and public welfare policies. Adequate growth and nutritional status of children are

monitored by the use of anthropometric measurements, specifically height and weight, which in

combination with the age of the child forms the anthropometric indices (Janevic et al., 2010).

Nutritional status is defined as “a person’s physiological level of nourishment in terms of energy

and protein stores, micronutrient status and metabolic functioning” (Kennedy, 2005). The

assessment of nutritional status is defined as “the science of determining nutrition status by

analyzing an individual’s medical, dietary, and social history; anthropometric data, biochemical

data, clinical data and drug-nutrient interactions” (Hammond et al., 2014, Janevic et al., 2010).

Thus, nutritional status is often used as a measure of social development. Furthermore, nutritional

status is strongly connected to health outcomes such as malnutrition (Masibo and Makoka, 2012).

Malnutrition is the condition that results from taking an unbalanced diet in which certain

nutrients are lacking, in excess (too high an intake), or in the wrong proportions (Dorland`s

Medical Dictionary). The term malnutrition therefore, refers to both under nutrition and over

nutrition. Under nutrition is the outcome of inadequate intake of nutrients. Under nutrition includes

being underweight for one’s age, too short for one’s age (stunted), dangerously thin (wasted), and

deficient in vitamins and minerals (Meshram et al., 2011). Over nutrition (obesity) is the outcome

24

of excess intake of nutrients. “For the purposes of this study, Malnutrition refers to under

nutrition”.

2.8 Weight for Age Status

Weight for age is used to measure a child’s weight in relation to his age (Furlong et al., 2016,

WHO, 2006). Underweight is defined as a weight for age below -2SD of the reference population,

while a weight for age of below -3SD of the reference population is classified as severe

underweight (Furlong et al., 2016, Bloem, 2007). Furthermore, classifications for assessing the

public health significance of malnutrition indicated that a prevalence of underweight that is less

than 10% indicates a low prevalence of malnutrition, whereas 10 to 19% indicates a medium

prevalence (Levin et al., 2016, Organization, 2016). In addition, 20 to 29% indicates a high

prevalence, while > 30% indicates a very high prevalence of underweight.

Nationally, 15% of children under age 5 are underweight (low weight-for-age), and 3% are

severely underweight. The proportion of underweight children is highest among those age 18-23

months (18%) (Jones et al., 2014). Regionally the national underweight prevalence is modest,

being lower than that observed in other countries (Botswana 11.2%, Egypt 7%, Ghana 11%,Kenya

11%, South Africa 8.7% and Zimbabwe 11.2%) and lower than some (Ethiopia 25.2%, Malawi

16.5%, Nigeria 19.8% and Niger 37.8%) (Jones et al., 2014, Organization, 2016, Demographic,

2015).

2.9 Height for Age Status

Height for age is a measure of how tall or short the child is relative to his age (Bloem, 2007,

WHO, 2006). Height does not increase rapidly in children and a low height for age reflects chronic

malnutrition, which is due to long-term starvation or shortage of food or repeated illness. Height

for age helps to identify children who are stunted or those who are very tall or above normal height.

25

Stunting is defined as a height for age of below -2SD of the reference population. In addition, a

height for age of below -3SD of the reference population is classified as severe stunting (Bloem,

2007, WHO, 2006).

Nationally, 40% of children under age 5 are stunted, and 17% are severely stunted. Analysis

by age groups shows that stunting is highest (54%) in children age 18-23 months and lowest (14%)

in children less than 6 months of age. Severe stunting shows a similar pattern, with the highest

proportion among children age 18-23 months (25%) (Jones et al., 2014). The stunting prevalence

of under-five children has been shown to be higher than many other African countries such as

Egypt having the stunting prevalence of 22.3%, Ghana 18.8%, Kenya 26.0%, Zimbabwe 27.6%

South Africa 22.0% and Nigeria 32.9%. Although other African countries have reported slightly

higher stunting prevalence compared to the national one of 40%, (Ethiopia 40.4%, DRC 42.6%,

Malawi 42.1%, Niger 43%), Zambia is ranked 116th

in the World and 10th

in Africa due to high

under five stunting prevalence levels (Organization, 2016, Demographic, 2015).

2.10 Mid-upper Arm Circumference (MUAC)

Mid-Upper Arm Circumference (MUAC) is the circumference of the left upper arm, measured

at the mid-point between the tip of the shoulder and the tip of the elbow. MUAC is used for the

assessment of nutritional status. The cut-offs commonly used are <11.5cm for severe acute

malnutrition, 11.5 to 12.4cm for moderate acute malnutrition, 12.5 to 13.4cm for risk of

malnutrition and > 13.5 as being well nourished. Using MUAC is simple and cheap, it is more

sensitive and is a better indicator of mortality risk associated with malnutrition than Weight-for-

Height. It is therefore used as surrogate measure of SAM than WAZ (Fiorentino et al., 2016, WHO,

2006).

26

2.11 Serum Vitamin A Status of Children

Micronutrient malnutrition, such as vitamin A and iron deficiency, is still a major public

health problem in developing countries. It was estimated that 163 million children in developing

countries were vitamin A deficient (Organization, 2009), Sanders et al., (2014) earlier estimated

that 140 million children younger than five years had vitamin A deficiency globally. It has also

been estimated that, globally, 127 million pre-school children were sub-clinically vitamin A

deficient (Sanders et al., 2014). Furthermore, nearly 100 million of those children with vitamin A

deficiency live in South Asia and sub-Saharan Africa (Sanders et al., 2014). In Sub- Saharan

Africa, 36 million pre-school children were affected by vitamin A deficiency (Sanders et al., 2014).

Although the extent of clinical vitamin A deficiency in South Africa is not as severe as it is in

some of the other sub-Saharan countries, one out of three children were identified as marginally

vitamin A deficient in the South Africa Vitamin A Consultative Group study (Labadarios et al.,

2008). Recent data indicate that two out of three children in South Africa have poor vitamin A

status (Labadarios et al., 2008). Nationally, not much has been documented concerning infant

serum vitamin A levels, but according to the estimates of Hotz and others, 56% of preschool aged

children are deficient in vitamin A (Hotz et al., 2012). In effort to reduce under 5 vitamin A

deficiency, Zambia has achieved high rates of vitamin A supplementation:96% of children 6–59

months of age receive the recommended two doses of vitamin approximately six month apart

(Organization, 2009).

2.12 Consequences of Under-nutrition on Mortality/Morbidity

Malnutrition in children is a global public health problem with wide implications.

Malnourished children have increased risk of dying from infectious diseases, and it is estimated

that malnutrition is the underlying cause of 45% of global deaths in children below 5 years of age

(Black et al., 2013). Although it has been debated whether malnutrition increases incidence of

27

infections, or whether it only increases severity of disease, solid data indicates that malnourished

children are at higher risk of dying once infected (Page et al., 2013). In a large analysis of data

derived from 10 prospective studies in Asia, Africa and South America, stunting, wasting and

underweight were each associated with excess mortality; even at Z-scores between -1 and -2

(Olofin et al., 2013). Other studies also reported malnutrition to be the reason for hospital

admissions in under five children for gastroenteritis (Mondal et al., 2009, Petri et al., 2008) and

Pneumonia (Olofin et al., 2013). Mondal also reported an increased risk to cryptosporidiosis,

amoebiasis and E. histolytica infections in Indian children (Mondal et al., 2009). Other studies

have also shown that poor nutritional status in children have a negative effect on brain and nervous

system development (Cusick and Georgieff, 2012, Martorell et al., 2009, Uauy et al., 2011). There

was a clear dose–response relationship between the degree of malnutrition and mortality, with the

risk among those with wasting greater than among those with stunting. Children with the most

profound anthropometric defects had an elevated risk of death from a variety of infections,

including sepsis, meningitis, measles and tuberculosis. Although some datasets in this analysis

were not adjusted for all potential confounders, and under nutrition may to some degree reflect

general socioeconomic disadvantage, nevertheless the body of evidence indicates that

malnourished children have a clear excess risk of infectious morbidity and mortality (Black et al.,

2013). Other specific nutritional deficiencies such as vitamin A deficiency (VAD) also increase

risk of death. In children, VAD results in increased risks of mortality and morbidity from measles

and diarrhoeal infections blindness, and anaemia (Black et al., 2013).

28

2.13 Malnutrition and the Immune System

Malnutrition has been described has the most common form of immunodeficiency and that

the increased susceptibility to infections may in part be caused by impairment of immune function

by malnutrition (Prendergast, 2015). The first-line defence of the immune system has been

considered to be the barrier function of the skin and mucosal surfaces, upheld by the physical

integrity of the epithelia, anti-microbial factors in secretions (e.g. lysozyme, secretory IgA and

gastric acidity) and the commensal bacterial flora. Studies have been conducted to describe the

barrier function in malnourished children, skin structure and function, structure and permeability

of intestinal mucosa, the protective factors in secretions and the microbial flora colonizing mucosal

surfaces. The skin barrier has mostly been studied in children with oedematous malnutrition, who

may develop a characteristic dermatosis, characterized by hyper-pigmentation, cracking and

scaling of the epidermis, resembling ‘‘peeling paint’’, providing a potential entry port for

pathogens (Heilskov et al., 2014). The intestinal mucosa of malnourished children has been studied

from as early as 1965 and investigators have described a thin-walled intestine in malnourished

children. Small-intestinal biopsies showed thinning of the mucosa (Korpe and Petri, 2012),

decrease in villous height (Ahmed et al., 2009, Fagbemi et al., 2008) altered villous morphology;

and infiltration of lymphocytes (Korpe and Petri, 2012). Electron-microscopy studies have shown

sparse brush border with shortened microvilli and sparse endoplasmic reticulum (Garrett et al.,

2010, Korpe and Petri, 2012). Such an intestine may predispose to bacterial and viral translocation,

making the malnourished infants more susceptible to infectious diseases. Two studies carried in

Gambia and Zambia, described immune cells in small intestinal biopsies from malnourished

children compared to well-nourished children and both reported increased lymphocyte infiltration,

more T-cells, and cells expressing HLA-DR in malnourished children compared to malnourished

children (Fagbemi et al., 2008). In a study to describe the colony in malnourished children,

29

investigators reported an increased vascularity, atrophy of the mucosa and a tendency to rectal

prolapse in children with oedematous malnutrition (Heilskov et al., 2014).

Vitamin A and its active metabolites have been shown to be involved in many steps of

protective immunity from the antigen uptake to Secretory IgA (SIgA) secretion in the lumen and

imprinting T and B cells with gut-homing receptors (Beijer et al., 2014, Mora et al., 2008, Pino-

Lagos et al., 2008). SIgA is the most abundant class of antibodies found in the human intestine

and it is considered as a first line of defence in protecting the intestinal epithelium from enteric

pathogens and toxins. Studies show that vitamin A-deficient persons may experience increased

susceptibility to a variety of pathogens including rotavirus due to decreased amounts of SIgA

(Zeng et al., 2013).

2.14 Malnutrition and Vaccination

The effects of malnutrition on vaccine performance, sero-conversion rates and serum

immunoglobulin titres have been the subject of extensive research for several decades (Gaayeb et

al., 2014) Studies show that malnourished children generally appear to respond adequately to

vaccines and other studies have conflicting results. However, the evidence base remains relatively

weak because most are small, old studies, using laboratory techniques and definitions of

malnutrition that are now outdated. The anthropometric indices of many of these studies were not

clearly defined neither was the time point of infant enrolment clearly indicated. There is, therefore,

an urgent need to better understand the fundamental immunology of malnutrition. Only a few of

such studies were carried out in Africa and the vaccines studied were not rotavirus vaccines.

Despite these limitations, it appears that malnourished children can generally mount protective

responses to protein (Deloria-Knoll et al., 2006), live (Savy et al., 2009), killed (Savy et al., 2009)

and polysaccharide vaccines (Savy et al., 2009, von Gottberg et al., 2014). To evaluate the effect

of nutritional status on the effectiveness of RV1 vaccine, a Meta–analysis was performed on the

efficacy studies conducted in Brazil, Mexico, and Venezuela. The results of this analysis showed

30

that malnutrition did not seem to influence efficacy of the RV1 vaccine. In this study, VE against

RVGE was similar among well-nourished and malnourished infants at 74% and 73% respectively

against severe RVGE and 61% for both against RVGE of any severity (Ruiz-Palacios et al., 2006).

In another study carried out in Brazil to evaluate the effectiveness of a different type of the

rotavirus vaccine RV5 in relation to infant nutritional status, it suggested that rotavirus vaccine

may achieve higher levels of protection among well-nourished than in malnourished infants (Savy

et al., 2009).Other studies did not directly look at association between nutritional status and

rotavirus vaccine effectiveness but rather looked at the possible association between the nutritional

status and rotavirus disease outcomes. A study carried out in Bangladesh to evaluate the impact of

nutritional status and Rotavirus diarrhoea found that better nutritional status among Bangladeshi

infants was strongly associated with higher risk of rotavirus diarrhea in the first three years of life.

This study had a limitation in that it failed to explain why well-nourished children were a more

risk of having RV-age than vice versa and secondly all diarrhea cases within the study cohort could

not be collected and analyzed suggesting that the results obtained were a not true indication of the

association in the population (Verkerke et al., 2016). Data regarding the potential impact of

malnutrition on rotavirus vaccines and other oral vaccines other than OPV are limited (Verkerke

et al., 2016). One of such studies conducted to determine sero-conversion rates after OPV

vaccination recorded significantly lower poliovirus antibody titers in conjunction with

underweight (weight-for-age Z score ≤2) after at least three doses of OPV (Haque et al., 2014).

However, other studies in India, Nigeria and Pakistan have not observed an association between

malnutrition and OPV immunogenicity (Haque et al., 2014).

Little work has been done in the context of malnutrition and rotavirus vaccines, in Zambia this

was the first study undertaken to study the relationship between nutritional status on rotavirus

vaccine sero-conversion in infants.

31

CHAPTER 3: METHODOLOGY

3.1 Study Design

This was a prospective cross-sectional study whose purpose was to evaluate if poor nutritional

status (vitamin A, weight for age, height for age and mid-upper arm circumference) affect the

ability of 6-12 weeks infants to sero-convert to rotavirus vaccination?

3.2 Study site

The study was conducted at Kamwala clinic, a peri-urban primary care health facility in Lusaka,

which provides basic outpatient and maternal child health services. Kamwala has a catchment

population of over 10,000 with approximately 120 new infants presenting each month. The

majority of the attendees at this public health facility are mostly low-income residents of two

nearby high density residential areas.

3.3 Study Population

Study recruitment was conducted between April 2013 and March 2014. 1320 Participants were

recruited as they presented to the facility for routine immunization services described in detail

previously and as described in figure 3 (Chilengi et al., 2016).

32

Figure 3. Study Participants Flow chart

3.3.1 Inclusion Criteria

Infants were eligible if: i) the mother was willing to participate voluntarily and able to provide

signed informed consent (with witness in the case of illiterate participant); ii) the infant was

eligible for RV1 vaccine immunization as per national policy (male or female infant, aged 6-12

weeks old); iii) the mother was willing to allow her infant to undergo study procedures, including

full-course RV1 vaccination, phlebotomy at enrolment and 1 month post full RV1 vaccination and

iv) planned to remain in the area and was willing to come for scheduled visits for the duration of

the study.

Total number

Screened (1320)

Infant Height for Age Status

(N= 208)

Infant Mid-Upper Arm circumference

(N= 208)

Serum Vitamin A concentration

(N= 208)

Enrolled

(N= 420)

Infant Weight for

Age Status

(N= 208)

Dropped due to exclusion

criteria and non-consent

(N= 900)

Dropped due to missing lab

report and/or insufficiTotal number Screened (1320)

Total number Screened

(1320)

ent sample (N= 212)

(N= 900)

Dropped due to missing lab

report and/or insufficient

sample (N= 212)

Infant serum Ig A Sero-conversion and final analysis

(N= 208)

33

3.3.2 Exclusion Criteria

Infants were not eligible for the study if they had received any rotavirus vaccine prior to the study,

suffered from any congenital or chronic gastrointestinal diseases including any uncorrected

congenital malformation of the gastrointestinal tract, received any immunosuppressant or

immunoglobulin, or had a history of gastroenteritis seven days prior to their visit to the clinic.

3.3.3 Sample Size Determination

Sample size calculation was achieved by using the population data which suggests a sero-

conversion rate of 60% (Chilengi et al., 2016) and a total of 232 infant paired samples were

required to detect 50% reduction in sero-conversion at 80% power with a 2 sided Chi-squared test

with a 5% level of significance.

3.3.3.1 Sampling Method

Simple random sampling was used to select participants/samples for analysis.

Sample size was established by the following formula;

Sample Size= 2SD2−(𝑍∝ + 𝑍1−𝛽)2

𝑑2

Where; SD is the Standard Deviation of the population =1.15,

Zα = 1.96 (From the Z table) at type 1 error of 5%, Z = 0.842 (from the Z table) at 80% power,

d = 0.30 expected effect size.

This gives us;

Sample Size =2*(1.15)2(1.96 + 0.842)

2/ (0.30)

2

Sample Size = 232 were required to detect the difference. Convenient sampling was used for there

was not enough samples to be included in the final analysis.

34

3.3.2 Clinic Procedures

Enrolled infants received a physical examination and a baseline serum sample was taken for

rotavirus-specific IgA antibodies to determine natural infection (serum anti RV-IgA sero-

positivity) and serum vitamin A concentration. Infants were then administered the full expanded

programme on immunization (EPI) vaccine regimen, which included the oral RV1 and

poliovaccines, and parenteral pentavalent vaccines (containing diphtheria, tetanus, pertussis,

haemophilus influenza, and hepatitis B). They were given an appointment card highlighting the

next scheduled visit (one month post the second RV dose) and were escorted home by a research

assistant in order to record the physical location of their residence, as there are no street addresses

in the clinic catchment area. The nutritional status of children was also assessed by anthropometric

measurements at base-line just before rotavirus vaccination. Child nutritional indicators of height

for age Z-scores (HAZ), weight for age Z scores (WAZ) and mid upper arm circumference

(MUAC) were calculated according to the WHO 2009 growth standard (WHO, 2006). We used

the most common thresholds: HAZ < −2 Z scores to define stunting or chronic malnutrition, WAZ

< −2 Z scores to define underweight or global malnutrition and MUAC of <11.5cm for severe

acute malnutrition, 11.5 to 12.4cm for moderate acute malnutrition, 12.5 to 13.4cm as risk of

malnutrition and > 13.5 as being well nourished. One month after the second dose of RV1 serum

sample was taken for rotavirus-specific IgA antibodies to determine infant sero-conversion

following vaccination.

35

Table 3.0 Z-score classification to determine nutritional status in children (WHO, 2006)

Z-score WAZ WHZ HAZ

Classification

< -3SD Severely underweight Severely wasted Severely stunted

-3SD to < -2SD Under weight Wasted Stunted

-2SD to < -1SD Mild under weight Mildly wasted Mildly stunted

-1SD to +1SD Normal weight Normal WHZ Normal height

>+1SD to ≤ +2SD Possible growth problem Possible risk of overweight Normal height

>+2SD to ≤ +3SD Possible growth problem Overweight Normal height

>+3SD Possible growth problem Obese Above normal

3.4 Laboratory Procedures.

3.4.1 Measurement of Serum IgA for the Determination of Sero-positivity and Sero-

conversion To measure infant serum IgA concentrations, ELISA methods were used at baseline to measure

serum IgA sero-positivity to determine infant natural rotavirus exposure and 1 month after the

second vaccine dose to assess sero-conversion as described elsewhere (Chilengi et al., 2016,

Groome et al., 2014). Sero-conversion was defined as a ≥ 4 fold rise in serum IgA titre from baseline

to one-month post RV1 dose 2, while sero-positivity of IgA was defined as serum titre ≥ 40.

36

3.4.2 Measurement of Serum Vitamin A to Determine the Prevalence of Vitamin A

Deficiency Serum samples were evaluated for Vitamin A level using Quantikine® ELISA human retinol

binding protein4 (RBP4) Immunoassay which is used as a surrogate measure for serum VA (RnS

systems, MN, USA) according to the manufacturer’s instructions. Vitamin A status was defined

as normal serum vitamin A concentration when serum RBP4 was > 0.70μmol/L, moderate deficiency

was defined as < 0.70μmol/L and severely deficient as ≤ 0.35 μmol/L of serum RBP4 concentration

respectively.

3.5 Data Analysis

Children were defined as having sero-converted when at 1 month post immunization, the rotavirus

specific-IgA titres increased by four fold or greater compared with the titre recorded before the

first dose of RV. Pearson Chi-squared test was used to investigate the association of sero-

conversion and categorical factors (i.e. sex, vitamin A status, underweight, stunting and infant

sero-positivity at baseline) at 95% confidence level and p value less than 0.05 was considered

statistically significant.

3.6 Ethical Considerations

Ethical Clearance was sought from the University of Zambia Biomedical Research Ethic

committee (UNZABREC) of the School of Medicine (REF. No. 013-02-16). The study posed no

additional risks to the participants as no extra samples were collected from them apart from those

indicated by the attending physician as part of routine standard of care. Patient names were not

used; instead, unique identification codes were used in order to ensure confidentiality. Written

consent was sought from parents/guardians of the participants prior to sample collection.

Information obtained from the patients was strictly confined to academic use only, unless

37

otherwise there was clinical indication necessary to allow a shared confidentiality in good faith of

the patient concerned.

3.7 Limitations

This study had several limitations, including the small sample size, and the lack of collection of

serum samples between doses. Only serum vitamin A was measured and other micronutrients like

Iron and Zinc whose deficiency have been implicated in diarrhoeal diseases and poor vaccine

response were not included in the study.

38

CHAPTER 4: RESULTS

4.1 Patient Recruitment and Descriptives

Of a total of 1320 infants that were screened, 420 were successfully enrolled. The most common

reason for declining to participate was unwillingness for the infant’s mother to have the infant’s

blood drawn (3.5ml of blood was drawn from each infant), methods of enrolment have been

published elsewhere (Chilengi et al., 2016). From the 420 infants who were successfully enrolled,

208 serum samples were analyzed to determine infant serum vitamin A concentration and serum

anti RV IgA at base-line and one month post vaccination.

4.2 Prevalence of Poor Nutritional Status in Infants and Infant Early Exposure to

Rotavirus Infection The nutritional status of the infants was determined by infant serum vitamin A status and weight

for age status, height for age status. Overall baseline serum vitamin A deficiency was 78.8%

(164/208). Baseline infant vitamin A moderate deficiency was 63.9%(133/208) and vitamin A

severe deficiency was14.9%(31/208). Infants who were categorized as being underweight or

stunted had Z-scores of ≤-2 and as having normal weight and height Z-scores of >-2 respectively.

Infant base-line underweight was 6.3%(13/208) and stunting was at 52.4%(109/208). MUAC

status was also used to determine infant nutritional status and those whose were well nourished

were 10.1%(21/208), 39.4%(82/208) of the infants were at risk of malnutrition, 37.95%(79/208)

of the infants had moderate acute malnutrition and 12.5% (26/208) infants had severe acute

malnutrition. Infant early exposure to rotavirus infection was determined by the presence of anti-

rotavirus specific IgA in infant serum at base-line and infant sero-positivity was 48(23.2%) as

shown in Table 4.1.

39

Table 4.1. The prevalence of poor nutritional status in infants attending routine immunization at Kamwala clinic

Characteristics Total (%N)

Sex (N=208)

Female 105(50.5)

Male 103(49.5)

Vitamin A status (N=208)

Normal 44(21.2)

Moderately deficient 133(63.9)

Severely deficient 31(14.9)

Underweight (N=208)

WAZ >-2 195(93.7)

WAZ≤-2 13(6.3)

Stunting (N=208)

HAZ >-2 99(47.6)

HAZ≤-2 109(52.4)

Mid-Upper Arm Circumference (MUAC)

(N=208)

>13.4 21(10.1)

12.5 - 13.4 82(39.4) 11.5 - 12.4 79(38.0)

<11.5 26(12.5)

Serum IgA sero-positivity (N=208)

positive 48(23.1)

negative 160(76.9) *Vitamin A status was defined as normal if serum RBP4 levels ˃ 0.7 μmol/l, moderate deficient if serum RBP4 levels ˂ 0.7 μmol/l and sevely deficient if serum RBP4 levels ≤ 0.35 μmol/l. Mid-Upper Arm Circumference(MUAC) was defined as well-nourished if MUAC ˃ 13.4 cm, at risk of

Malnutrition if MUAC = 12.5-13.4cm, moderate acute malnutrition if MUAC = 11.5-12.4cm and severe acute malnutrition if ˂ 11.5cm.. Seropositivity of

IgA was defined as titer ≥1:40. Weight was defined as normal if WAZ ˃ -2 and underweight if WAZ ≤ -2. Stunting was defined as normal if HAZ ˃ -2

and stunted if HAZ ≤ -2

4.3 Effects of Nutritional Status on Sero-conversion Among Children 3 months in Lusaka

Zambia After one month following the second dose of rotavirus vaccination infant serum IgA sero-conversion

rates were determined and the overall sero-conversion frequency was 57.2% (119/208). Pearson Chi-

squared test was then used to investigate the association of sero-conversion and categorical factors (i.e.

sex, vitamin A status, underweight, stunting and mid-upper arm circumference at baseline). The sero-

conversion rate for the females was at 57.1% and 57.3% for the males. The sero-conversion rates for

severely vitamin A deficient infants was 61.3%, 56.4% for moderately deficient and 56.8% for the

vitamin A sufficient infants. The sero-conversion rate of the infants was not significantly affected by the

vitamin A status of the infants (p= 0.882). Underweight and stunted infants who sero-converted were

53.9% and 56.9% respectively. Being underweight or stunted did not significantly affect the ability of

the infants to sero-convert to the rotavirus vaccine. The sero-conversion rates of infants who were well

40

nourished as determined by their MUAC was 13(61.9%), 50(61.0%) for those who at were risk of

malnutrition, 44(55.70%) for those who were moderately malnourished and for the infants who were

severely malnourished was 12(46.2). Moderate or severe acute malnutrition did not significantly affect

the ability of these infants to sero-convert to rotavirus vaccine (p=0.565). Infant who were exposed to

rotavirus infection before RV1 vaccine administration had a sero-conversion rate of 28(48.3%) and early

exposure to rotavirus infection did not significantly affect the ability of these infants to sero-convert to

rotavirus vaccine (p=0.832) as shown in Table 4.2.

41

Table 4.2. Effects of nutritional status on sero-conversion among children 3 months in Lusaka Zambia

Number (%)

Characteristics sero-conversion 95%CI P-value

Sex (N=208) 0.984

Female 60(57.1) [47.43, 66.34]

Male 59(57.3) [47.47, 66.55]

Vitamin A status (N=208) 0.882

Severely deficient 19(61.3) [43.06,76.83]

Moderately deficient 75(56.4) [47.78, 64.64]

Normal 25(56.8) [41.76, 70.71]

Underweight (N=208) 0.812

WAZ>-2 111(57.2) [50.09, 64.04]

WAZ≤-2 7(53.8) [27.14, 78.51]

Stunting (N=208) 0.919

HAZ >-2 57(57.6) [47.56, 67.00]

HAZ≤-2 62(56.9) [47.35 , 65.93]

Mid-Upper Arm Circumference

(MUAC) (N=208) 0.565

>13.4 13 (61.9) [40.50, 83.31]

12.5 - 13.4 50 (61.0) [50.29, 71.66]

11.5 - 12.4 44 (55.7) [44.61, 66.78]

<11.4 12 (46.2) [26.50, 65.81]

IgA Seropositivitity (N=208) 0.832

Positive 28(48.33) [43.86,76.50]

Negative 90(56.60) [48.73,64.16]

* seroconversion = post immunization rotavirus specific-IgA titre increased ≥four-fold compared with the titre recorded before the first dose

of RV1

42

CHAPTER 5: DISCUSSION, CONCLUSION AND RECOMENDATIONS

5.1 Discussion

The main focus of this study was to investigate if the nutritional status (Vitamin A, WAZ, HAZ

and MUAC) have an effect on the sero-conversion following rotavirus vaccination. There were

four key findings from this study: a) Infant serum specific RV-IgA sero-conversion after RV

vaccination was not influenced by the base-line VA status of the infants although VAD was

generally high in this cohort. b) base-line infant underweight was low and that underweight did

not influence the level of Infant serum specific anti RV-IgA sero-conversion rate following RV

vaccination; c) base-line infant stunting was very high and that stunting did not influence the level

of Infant serum specific RV-IgA sero-conversion rate following RV vaccination; d) base-line

severe acute malnutrition determined by the infant mid-upper arm circumference (MUAC) did not

significantly affect infant sero-conversion rates following RV vaccination.

The effect of VA status and malnutrition on vaccine responses in children in recent times has

received particular attention, although their effect on rotavirus vaccines has not been extensively

studied (Heilskov et al., 2014, Savy et al., 2009). Our findings of no association between VA status

and rotavirus IgA sero-conversion rates have been consistent with the results of randomised

controlled trials, observational and cross-sectional studies on the effects of vitamin A

supplementation (VAS) and VAD in children on sero-conversion rates of vaccines. VAS was

shown to be associated with a small decrease in response to the tuberculin skin test after BCG

vaccination in children from Malawi and Guinea-Bissau respectively (Diness et al., 2007). It has

also being shown that although mean antibody titters tended to be higher in participants who

received VA, sero-conversion rates were universally high and did not significantly differ between

supplemented and un-supplemented participants in measles vaccine (MV) in studies carried out in

43

India, Jamaica and Guinea-Bissau respectively (Benn et al., 2002, Rytter et al., 2014). VA has

been shown to have no effect on sero-conversion rates to any of the 3 serotypes of poliomyelitis

vaccine, (Bahl et al., 2002) and has no discernable effect on other vaccines, including,

cholera,(Qadri et al., 2016) influenza (Gardner et al., 2000), Hib (Zandoh et al., 2007), and

pneumococcal vaccines (Deloria-Knoll et al., 2006).

Our observed low prevalence of under-weight and high prevalence of stunting was in-line with the

report of WHO on global nutritional trends 2016 (Organization, 2016), this report suggested that

some countries have low underweight prevalence but unacceptably high stunting rates, for

example, in Guatemala, Liberia, Malawi, Mozambique, Rwanda, and the United Republic of

Tanzania, child underweight prevalence is lower than 20 per cent, while stunting prevalence

remains above 40%. Our infant underweight prevalence was also in line with the findings of the

Zambia Demographic of Health Survey of 2013- 2014 (Demographic, 2015) but stunting rates are

higher in our observation ((51.2% (103/201)) than the predicted rates of the ZDHS for Lusaka

Province stunting rates (35.7%) (Demographic, 2015). Malnutrition defined as low HAZ and WAZ

was shown to have no detectable effect on antibody responses to other vaccines, like the measles

vaccine (Bhaskaram, 1995). A study carried out in Nigeria to assess the effect of malnutrition on

responses to OPV in young children, found that the mean increase in antibody titres after 3 doses

of OPV did not significantly differ between infants severely or moderately malnourished

(according to their weight-for-age) and well-nourished infants (Greenwood et al., 1986). In

addition, no correlation was found between the antibody response to OPV and serum albumin

levels, used as an indicator of nutritional status. Similarly, a study conducted in 62 Indian children

aged 1–12 mo showed no difference in sero-conversion rates after 5 doses of the trivalent OPV

between under-weight and non-underweight children (L Heymann et al., 1987). However, the

analyses were not adjusted for any variables, not even the pre-vaccination antibody levels, which

44

were found to be sero-positive for some of the children. These findings were not in agreement with

an Egyptian study that was conducted in 11- to 23-mo-old children (Hafez et al., 1977). Children

suffering from kwashiorkor and marasmus were matched with healthy children of the same age

and sex and they all received a single dose of the OPV. The authors reported that antibody titers

to the vaccine were significantly lower in children with kwashiorkor or marasmus than normal

controls, suggesting a depression in antibody production against poliovirus vaccine in children

with malnutrition. A study carried out in Brazil to evaluate the effects of malnutrition of the sero-

conversion rates to RRV-TV infants, suggested that RRV-TV may achieve higher levels of ser-

conversion among well-nourished than in malnourished infants (Linhares et al., 2002). On the

MUAC basis, a study carried out in the Gambia by Moore (Moore et al., 2003) found no significant

association between MUAC indices and sero-conversion rates following the double doses of rabies

vaccines. The findings of these studies are not all in agreement with the findings of our study,

although similarities where seen in the findings of the Nigerian and Indian studies (Greenwood et

al., 1986, Hafez et al., 1977), the study populations for these studies were quiet small, the effects

of natural infections were over-looked and the studies were done over 30 ago using rather old

methods. The age of the study participants were usually older than the cohort in our study, 6-12

weeks old infants. This age group is important for such studies because it is at this age that most

vaccines are recommended for administration including OPV, rotavirus vaccines, and parenteral

pentavalent vaccines (containing diphtheria, tetanus, pertussis, haemophilus influenza, and

hepatitis B) therefore, understanding the nutritional status of the infants just before vaccination

would better the understanding of the performance of vaccines in relation to their nutritional status.

45

Another important finding our study was the relatively high rate of pre-vaccination IgA sero-

positivity. This finding suggests that almost quarter of the infants were already exposed to wild

rotavirus infection very early in life before rotavirus vaccination. There was no evidence of

association between infant serum IgA sero-conversion and serum IgA sero-positivity, it remains

to be concluded whether early natural exposure could also negatively influence vaccine sero-

conversion as previously observed in South Africa and in Nicaragua (Becker-Dreps et al., 2014,

Groome et al., 2014).

Our observation of no evidence of association between infant nutritional status (vitamin A status,

HAZ, WAZ and MUAC) was not always consistent with the findings of most of the studies

conducted on malnutrition and vaccination and we therefore, suggest more research in this light to

investigate the particular arm of the immune response poor infant nutritional status may have on

the rotavirus vaccine (Prendergast, 2015, Savy et al., 2009). As many LMICs seek to attain

millennium development goals, reduction of child mortality through interventions that include

“effective child vaccines” is an important strategy. Research such as ours, seeking to understand

and explain challenges of achieving rotavirus vaccine impact is of great importance.

5.2 Conclusion

The complexity and lack of understanding of sero-conversion to vaccines and its meaning in terms

of clinical protection or vaccine efficacy still prevails at this point. Based on our findings, we

conclude that poor IgA sero-conversion frequency observed in this cohort was not influenced by

nutritional factors indicated by infant serum Vitamin A, HAZ, WAZ and MUAC. These factors

may have other effects later in childhood, as no difference was observed among sero-convertors

and non-sero-convertors to rotavirus vaccination in the 6-12 weeks age group.

46

5.3 Recommendations

Further research is needed to better understand vaccine sero-conversion. The relationship between

EED and RV vaccine efficacy remains a subject of interest for our on-going research.

47

REFERENCES

Agócs, M. M., Serhan, F., Yen, C., Mwenda, J. M., De Oliveira, L. H., Teleb, N., Wasley, A.,

Wijesinghe, P. R., Fox, K. & Tate, J. E. 2014. WHO global rotavirus surveillance network:

a strategic review of the first 5 years, 2008–2012. MMWR. Morbidity and mortality weekly

report, 63, 634.

Ahmed, T., Arifuzzaman, M., Lebens, M., Qadri, F. & Lundgren, A. 2009. CD4+ T-cell responses

to an oral inactivated cholera vaccine in young children in a cholera endemic country and

the enhancing effect of zinc supplementation. Vaccine, 28, 422-9.

Aiyegbo, M. S., Sapparapu, G., Spiller, B. W., Eli, I. M., Williams, D. R., Kim, R., Lee, D. E.,

Liu, T., Li, S., Woods, V. L., Jr., Nannemann, D. P., Meiler, J., Stewart, P. L. & Crowe, J.

E., Jr. 2013. Human rotavirus VP6-specific antibodies mediate intracellular neutralization

by binding to a quaternary structure in the transcriptional pore. PLoS One, 8, e61101.

Angel, J., Franco, M. A. & Greenberg, H. B. 2012. Rotavirus immune responses and correlates of

protection. Current Opinion in Virology, 2, 419-25.

Appaiahgari, M. B., Glass, R., Singh, S., Taneja, S., Rongsen-Chandola, T., Bhandari, N., Mishra,

S. & Vrati, S. 2014. Transplacental rotavirus IgG interferes with immune response to live

oral rotavirus vaccine ORV-116E in Indian infants. Vaccine, 32, 651-6.

Armah, G., Pringle, K., Enweronu- Laryea, C. C., Ansong, D., Mwenda, J. M., Diamenu, S. K.,

Narh, C., Lartey, B., Binka, F., Grytdal, S., Patel, M., Parashar, U. & Lopman, B. 2016.

Impact and Effectiveness of Monovalent Rotavirus Vaccine Against Severe Rotavirus

Diarrhea in Ghana. Clinical Infectious Diseases, 62 Suppl 2, S200-7.

Armah, G. E. & Binka, F. N. 2014. Sustaining rotavirus vaccination in Africa: measuring vaccine

effectiveness. Lancet Infectious Diseases, 14, 1031-1032.

Armah, G. E., Sow, S. O., Breiman, R. F., Dallas, M. J., Tapia, M. D., Feikin, D. R., Binka, F. N.,

Steele, A. D., Laserson, K. F., Ansah, N. A., Levine, M. M., Lewis, K., Coia, M. L., Attah-

Poku, M., Ojwando, J., Rivers, S. B., Victor, J. C., Nyambane, G., Hodgson, A., Schodel,

F., Ciarlet, M. & Neuzil, K. M. 2010. Efficacy of pentavalent rotavirus vaccine against

severe rotavirus gastroenteritis in infants in developing countries in sub- Saharan Africa: a

randomised, double-blind, placebo-controlled trial. Lancet, 376, 606-14.

Aupiais, C., De Rougemont, A., Menager, C., Vallet, C., Brasme, J. F., Kaplon, J., Pothier, P. &

Gendrel, D. 2009. Severity of acute gastroenteritis in infants infected by G1 or G9

rotaviruses. Journal of Clinical Virology, 46, 282-5.

Bahl, R., Bhandari, N., Kant, S., Molbak, K., Ostergaard, E. & Bhan, M. K. 2002. Effect of vitamin

A administered at Expanded Program on Immunization contacts on antibody response to

oral polio vaccine. European Journal of Clinical Nutrition, 56, 321-5.

48

Becker-Dreps, S., Bucardo, F., Vilchez, S., Zambrana, L. E., Liu, L., Weber, D. J., Peña, R.,

Barclay, L., Vinjé, J., Hudgens, M. G., Nordgren, J., Svensson, L., Morgan, D. R.,

Espinoza, F. & Paniagua, M. 2014. Etiology of childhood diarrhea after rotavirus vaccine

introduction: a prospective, population-based study in Nicaragua. The Pediatric infectious

disease journal, 33, 1156-1163.

Beijer, M. R., Kraal, G. & Den Haan, J. M. 2014. Vitamin A and dendritic cell differentiation.

Immunology, 142, 39-45.

Benn, C. S., Balde, A., George, E., Kidd, M., Whittle, H., Lisse, I. M. & Aaby, P. 2002. Effect of

vitamin A supplementation on measles-specific antibody levels in Guinea-Bissau. Lancet,

359, 1313-4.

Beres, L. K., Tate, J. E., Njobvu, L., Chibwe, B., Rudd, C., Guffey, M. B., Stringer, J. S., Parashar,

U. D. & Chilengi, R. 2016. A Preliminary Assessment of Rotavirus Vaccine Effectiveness

in Zambia. Clinical Infectious Diseases, 62 Suppl 2, S175-82.

Bhandari, N., Rongsen-Chandola, T., Bavdekar, A., John, J., Antony, K., Taneja, S., Goyal, N.,

Kawade, A., Kang, G., Rathore, S. S., Juvekar, S., Muliyil, J., Arya, A., Shaikh, H.,

Abraham, V., Vrati, S., Proschan, M., Kohberger, R., Thiry, G., Glass, R., Greenberg, H.

B., Curlin, G., Mohan, K., Harshavardhan, G. V., Prasad, S., Rao, T. S., Boslego, J. &

Bhan, M. K. 2014. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in

Indian children in the second year of life. Vaccine, 32 Suppl 1, A110-6.

Bhaskaram, P. 1995. Measles & malnutrition.

Bhowmick, R., Halder, U. C., Chattopadhyay, S., Nayak, M. K. & Chawla-Sarkar, M. 2013.

Rotavirus-encoded nonstructural protein 1 modulates cellular apoptotic machinery by

targeting tumor suppressor protein p53. Journal of virology, 87, 6840-6850.

Black, R. E., Victora, C. G., Walker, S. P., Bhutta, Z. A., Christian, P., De Onis, M., Ezzati, M.,

Grantham-Mcgregor, S., Katz, J., Martorell, R. & Uauy, R. 2013. Maternal and child

undernutrition and overweight in low-income and middle-income countries. Lancet, 382,

427-451.

Block, S. L., Vesikari, T., Goveia, M. G., Rivers, S. B., Adeyi, B. A., Dallas, M. J., Bauder, J.,

Boslego, J. W. & Heaton, P. M. 2007. Efficacy, immunogenicity, and safety of a

pentavalent human-bovine (WC3) reassortant rotavirus vaccine at the end of shelf life.

Pediatrics, 119, 11-8.

Bloem, M. 2007. The 2006 WHO child growth standards. BMJ (Clinical research ed.), 334, 705-

706.

Blutt, S. E., Matson, D. O., Crawford, S. E., Staat, M. A., Azimi, P., Bennett, B. L., Piedra, P. A.

& Conner, M. E. 2007. Rotavirus antigenemia in children is associated with viremia.

PLoS Med, 4, e121.

49

Boom, J. A., Tate, J. E., Sahni, L. C., Rench, M. A., Hull, J. J., Gentsch, J. R., Patel, M. M., Baker,

C. J. & Parashar, U. D. 2010. Effectiveness of pentavalent rotavirus vaccine in a large

urban population in the United States. Pediatrics, 125, e199-207.

Buttery, J. P., Lambert, S. B., Grimwood, K., Nissen, M. D., Field, E. J., Macartney, K. K.,

Akikusa, J. D., Kelly, J. J. & Kirkwood, C. D. 2011. Reduction in rotavirus-associated

acute gastroenteritis following introduction of rotavirus vaccine into Australia's National

Childhood vaccine schedule. The Pediatric infectious disease journal, 30, S25-9.

Chilengi, R., Simuyandi, M., Beach, L., K. Becker-Dreps S, M., Emperador, D., Velasquez, D.,

Bosomprah, S. & Jiang, B. 2016. Association of Maternal Immunity with Rotavirus Vaccine

Immunogenicity in Zambian Infants.

Clarke, E. & Desselberger, U. 2014. Correlates of protection against human rotavirus disease and

the factors influencing protection in low-income settings. Mucosal Immunology, 8, 1.

Corcoran, M., Well, G. & Loo, I. 2014. Diagnosis of viral gastroenteritis in children:

interpretation of real-time PCR results and relation to clinical symptoms.

Cortese, M. M., Immergluck, L. C., Held, M., Jain, S., Chan, T., Grizas, A. P., Khizer, S., Barrett,

C., Quaye, O., Mijatovic-Rustempasic, S., Gautam, R., Bowen, M. D., Moore, J., Tate, J.

E., Parashar, U. D. & Vazquez, M. 2013. Effectiveness of monovalent and pentavalent

rotavirus vaccine. Pediatrics, 132, e25-33.

Corthesy, B., Benureau, Y., Perrier, C., Fourgeux, C., Parez, N., Greenberg, H. & Schwartz-Cornil,

I. 2006. Rotavirus anti- VP6 secretory immunoglobulin A contributes to protection via

intracellular neutralization but not via immune exclusion. Journal of Virology, 80, 10692-

9.

Criglar, J. M., Hu, L., Crawford, S. E., Hyser, J. M., Broughman, J. R., Prasad, B. V. & Estes, M.

K. 2014. A novel form of rotavirus NSP2 and phosphorylation-dependent NSP2-NSP5

interactions are associated with viroplasm assembly. Journal of Virology, 88, 786-98.

Cusick, S. E. & Georgieff, M. K. 2012. Nutrient supplementation and neurodevelopment: timing

is the key. Archives of Pediatrics and Adolescent Medicine, 166, 481-2.

Danchin, M. H. & Bines, J. E. 2009. Defeating rotavirus? The global recommendation for rotavirus

vaccination. New England Journal of Medicine, 361, 1919-1921.

Deal, E. M., Lahl, K., Narvaez, C. F., Butcher, E. C. & Greenberg, H. B. 2013. Plasmacytoid

dendritic cells promote rotavirus-induced human and murine B cell responses. Journal of

Clinical Investigations, 123, 2464-74.

Deloria-Knoll, M., Steinhoff, M., Semba, R. D., Nelson, K., Vlahov, D. & Meinert, C. L. 2006.

Effect of zinc and vitamin A supplementation on antibody responses to a pneumococcal

conjugate vaccine in HIV-positive injection drug users: a randomized trial. Vaccine, 24,

1670-9.

Demographic, Z. 2015. Health survey (ZDHS) 2013-2014. House hold population by Age and sex.

Lusaka: Zambia.

50

Dennehy, P. H. 2008. Rotavirus Vaccines: an Overview. Clinical Microbiology Reviews, 21, 198-

208.

Desselberger, U. 2014. Rotaviruses. Virus Research, 190, 75-96.

Diness, B. R., Fisker, A. B., Roth, A., Yazdanbakhsh, M., Sartono, E., Whittle, H., Nante, J. E.,

Lisse, I. M., Ravn, H., Rodrigues, A., Aaby, P. & Benn, C. S. 2007. Effect of high-dose

vitamin A supplementation on the immune response to Bacille Calmette-Guerin vaccine.

American Journal of Clinical Nutrition, 86, 1152-9.

Dormitzer, P. R., Nason, E. B., Prasad, B. V. & Harrison, S. C. 2004. Structural rearrangements

in the membrane penetration protein of a non-enveloped virus. Nature, 430, 1053-8.

Fagbemi, A. O., Amadi, B., Salvestrini, C., Eklund, E. A., Torrente, F., Freeze, H. H., Golden, M.

H., Mwiya, M., Kelly, P., Day, R. & Murch, S. H. 2008. Reduced production of sulfated

glycosaminoglycans occurs in Zambian children with kwashiorkor but not marasmus. The

American Journal of Clinical Nutrition, 89, 592-600.

Feng, N., Sen, A., Nguyen, H., Vo, P., Hoshino, Y., Deal, E. M. & Greenberg, H. B. 2009.

Variation in Antagonism of the Interferon Response to Rotavirus NSP1 Results in

Differential Infectivity in Mouse Embryonic Fibroblasts. Journal of Virology, 83, 6987-

6994.

Fiorentino, M., Sophonneary, P., Laillou, A., Whitney, S., De Groot, R., Perignon, M., Kuong, K.,

Berger, J. & Wieringa, F. T. 2016. Current MUAC Cut-Offs to Screen for Acute

Malnutrition Need to Be Adapted to Gender and Age: The Example of Cambodia. PloS

one, 11, e0146442-e0146442.

Fischer, T. K., Valentiner-Branth, P., Steinsland, H., Perch, M., Santos, G., Aaby, P., Mølbak, K.

& Sommerfelt, H. 2002. Protective Immunity after Natural Rotavirus Infection: A

Community Cohort Study of Newborn Children in Guinea-Bissau, West Africa. The

Journal of Infectious Diseases, 186, 593-597.

Franco, M. A., Angel, J. & Greenberg, H. B. 2006a. Immunity and correlates of protection for

rotavirus vaccines. Vaccine, 24, 2718-2731.

Franco, M. A., Angel, J. & Greenberg, H. B. 2006b. Immunity and correlates of protection for

rotavirus vaccines. Vaccine, 24, 2718-31.

Furlong, K. R., Anderson, L. N., Kang, H., Lebovic, G., Parkin, P. C., Maguire, J. L., O'connor, D. L. & Birken, C. S. 2016. BMI-for-Age and Weight-for-Length in Children 0 to 2 Years. Pediatrics, 138.

Gaayeb, L., Pincon, C., Cames, C., Sarr, J. B., Seck, M., Schacht, A. M., Remoue, F., Hermann,

E. & Riveau, G. 2014. Immune response to Bordetella pertussis is associated with season

and undernutrition in Senegalese children. Vaccine, 32, 3431-7.

51

Gardner, E. M., Bernstein, E. D., Popoff, K. A., Abrutyn, E., Gross, P. & Murasko, D. M. 2000.

Immune response to influenza vaccine in healthy elderly: lack of association with plasma

beta-carotene, retinol, alpha-tocopherol, or zinc. Mechanism of Ageing Development

Journal, 117, 29-45.

Garrett, W. S., Gordon, J. I. & Glimcher, L. H. 2010. Homeostasis and inflammation in the

intestine. Cell, 140, 859-70.

Giaquinto, C., Van Damme, P., Huet, F., Gothefors, L., Maxwell, M., Todd, P. & Da Dalt, L. 2007.

Clinical consequences of rotavirus acute gastroenteritis in Europe, 2004-2005: the

REVEAL study. Journal of Infectious Diseases, 195 Suppl 1, S26-35.

Glass, R. I., Parashar, U. D., Bresee, J. S., Turcios, R., Fischer, T. K., Widdowson, M. A., Jiang,

B. & Gentsch, J. R. 2006. Rotavirus vaccines: current prospects and future challenges.

Lancet, 368, 323-32.

Gray, J., Vesikari, T., Van Damme, P., Giaquinto, C., Mrukowicz, J., Guarino, A., Dagan, R.,

Szajewska, H. & Usonis, V. 2008. Rotavirus. Journal of Pediatric Gastroenterology and

Nutrition, 46, S24-S31.

Greenberg, H. B. 2011. Rotavirus vaccination and intussusception--act two. New England Journal

of Medicine, 364, 2354-5.

Greenberg, H. B. & Estes, M. K. 2009. Rotaviruses: from pathogenesis to vaccination.

Gastroenterology, 136, 1939-51.

Greenwood, B. M., Bradley-Moore, A. M., Bradley, A. K., Kirkwood, B. R. & Gilles, H. M. 1986.

The immune response to vaccination in undernourished and well-nourished Nigerian

children. Annals of Tropical Medicine & Parasitology, 80, 537-544.

Groome, M. J., Page, N., Cortese, M. M., Moyes, J., Zar, H. J., Kapongo, C. N., Mulligan, C.,

Diedericks, R., Cohen, C., Fleming, J. A., Seheri, M., Mphahlele, J., Walaza, S., Kahn, K.,

Chhagan, M., Steele, A. D., Parashar, U. D., Zell, E. R. & Madhi, S. A. 2014. Effectiveness

of monovalent human rotavirus vaccine against admission to hospital for acute rotavirus

diarrhoea in South African children: a case-control study. The Lancet Infectious Diseases,

14, 1096-1104.

Haber, P., Parashar, U. D., Haber, M. & Destefano, F. 2015. Intussusception after monovalent

rotavirus vaccine-United States, Vaccine Adverse Event Reporting System (VAERS),

2008-2014. Vaccine, 33, 4873-7.

Haber, P., Patel, M., Izurieta, H. S., Baggs, J., Gargiullo, P., Weintraub, E., Cortese, M., Braun,

M. M., Belongia, E. A., Miller, E., Ball, R., Iskander, J. & Parashar, U. D. 2008.

Postlicensure monitoring of intussusception after RotaTeq vaccination in the United

States, February 1, 2006, to September 25, 2007. Pediatrics, 121, 1206-12.

Hafez, M., Aref, G. H., Mehareb, S. W., Kassem, A. S., El-Tahhan, H., Rizk, Z., Mahfouz, R. &

Saad, K. 1977. Antibody production and complement system in protein energy

malnutrition. The Journal of tropical medicine and hygiene, 80, 36-39.

52

Hammond, M. I., Myers, E. F. & Trostler, N. 2014. Nutrition Care Process and Model: An

Academic and Practice Odyssey. Journal of the Academy of Nutrition and Dietetics, 114,

1879-1891.

Haque, R., Snider, C., Liu, Y., Ma, J. Z., Liu, L., Nayak, U., Mychaleckyj, J. C., Korpe, P., Mondal,

D., Kabir, M., Alam, M., Pallansch, M., Oberste, M. S., Weldon, W., Kirkpatrick, B. D. &

Petri, W. A., Jr. 2014. Oral polio vaccine response in breast fed infants with malnutrition

and diarrhea. Vaccine, 32, 478-482.

Hashizume, M., Armstrong, B., Wagatsuma, Y., Faruque, A. S., Hayashi, T. & Sack, D. A. 2008. Rotavirus infections and climate variability in Dhaka, Bangladesh: a time-series analysis. Epidemiology and Infections, 136, 1281-9.

Heilskov, S., Rytter, M. J. H., Vestergaard, C., Briend, A., Babirekere, E. & Deleuran, M. S. 2014.

Dermatosis in children with oedematous malnutrition (Kwashiorkor): a review of the

literature. Journal of the European Academy of Dermatology and Venereology, 28, 995-

1001.

Hemming, M. & Vesikari, T. 2014. Detection of rotateq vaccine-derived, double-reassortant

rotavirus in a 7-year- old child with acute gastroenteritis. The Pediatric infectious disease

journal, 33, 655-656.

Hotz, C., Chileshe, J., Siamusantu, W., Palaniappan, U. & Kafwembe, E. 2012. Vitamin A intake

and infection are associated with plasma retinol among pre-school children in rural Zambia.

Public Health and Nutrition, 15, 1688-96.

Hou, Z., Huang, Y., Huan, Y., Pang, W., Meng, M., Wang, P., Yang, M., Jiang, L., Cao, X. & Wu,

K. K. 2008. Anti-NSP4 antibody can block rotavirus-induced diarrhea in mice. Journal of

pediatric gastroenterology and nutrition, 46, 376-385.

Hu, L., Crawford, S. E., Hyser, J. M., Estes, M. K. & Prasad, B. V. 2012. Rotavirus non-

structural proteins: structure and function. Current Opinions in Virology, 2, 380-8.

Janevic, T., Borrell, L. N., Savitz, D. A., Herring, A. H. & Rundle, A. 2010. Neighbourhood food

environment and gestational diabetes in New York City. Paediatric and Perinatal

Epidemiology, 24, 249-254.

Jiang, V., Jiang, B., Tate, J., Parashar, U. D. & Patel, M. M. 2010. Performance of rotavirus

vaccines in developed and developing countries. Human Vaccines, 6, 532-42.

Johansson, E., Istrate, C., Charpilienne, A., Cohen, J., Hinkula, J., Poncet, D., Svensson, L. &

Johansen, K. 2008. Amount of maternal rotavirus-specific antibodies influence the

outcome of rotavirus vaccination of newborn mice with virus-like particles. Vaccine, 26,

778-85.

Jones, A. D., Ickes, S. B., Smith, L. E., Mbuya, M. N., Chasekwa, B., Heidkamp, R. A., Menon,

P., Zongrone, A. A. & Stoltzfus, R. J. 2014. World Health Organization infant and young

child feeding indicators and their associations with child anthropometry: a synthesis of

recent findings. Maternal Child Nutrition, 10, 1-17.

53

Kawamura, N., Tokoeda, Y., Oshima, M., Okahata, H., Tsutsumi, H., Van Doorn, L. J., Muto, H.,

Smolenov, I., Suryakiran, P. V. & Han, H. H. 2011. Efficacy, safety and immunogenicity

of RIX4414 in Japanese infants during the first two years of life. Vaccine, 29, 6335-41.

Kennedy, E. T. 2005. The global face of nutrition: what can governments and industry do? The

Journal of nutrition, 135, 913-915.

Kim, I. S., Trask, S. D., Babyonyshev, M., Dormitzer, P. R. & Harrison, S. C. 2010. Effect of

mutations in VP5 hydrophobic loops on rotavirus cell entry. Journal of Virology, 84, 6200-

7.

Korpe, P. S. & Petri, W. A., Jr. 2012. Environmental enteropathy: critical implications of a poorly

understood condition. Trends in Molecular Medicine, 18, 328-36.

L Heymann, D., Murphy, K., Brigaud, M., Aymard, M., Tembon, A. & K Maben, G. 1987. Oral

poliovirus vaccine in tropical Africa: Greater impact on incidence of paralytic disease than

expected from coverage surveys and seroconversion rates.

Labadarios, D., Swart, E. & Maunder, E. 2008. Executive summary of the National Food

Consumption Survey Fortification Baseline (NFCS-FB-I) South Africa, 2005.

Lee, R. M., Lessler, J., Lee, R. A., Rudolph, K. E., Reich, N. G., Perl, T. M. & Cummings, D. a.

T. 2013. Incubation periods of viral gastroenteritis: a systematic review. BMC Infectious

Diseases, 13, 446.

Leshem, E., Lopman, B., Glass, R., Gentsch, J., Bányai, K., Parashar, U. & Patel, M. 2014.

Distribution of rotavirus strains and strain-specific effectiveness of the rotavirus vaccine

after its introduction: a systematic review and meta-analysis. The Lancet Infectious

Diseases, 14, 847-856.

Levin, D., Marryat, L., Cole, T. J., Mccoll, J., Harjunmaa, U., Ashorn, P. & Wright, C. 2016. Fit

to WHO weight standard of European infants over time. Archives of Disease in Childhood,

101, 455-60.

Linhares, A. C., Carmo, K. B. D., Oliveira, K. K., Oliveira, C. S., Freitas, R. B. D., Bellesi, N.,

Monteiro, T. a. F., Gabbay, Y. B. & Mascarenhas, J. D. a. P. 2002. Nutritional status in

relation to the efficacy of the rhesus-human reassortant, tetravalent rotavirus vaccine

(RRV-TV) in infants from Belém, pará state, Brazil. Revista do Instituto de Medicina

Tropical de Sao Paulo, 44, 13-16.

Linhares, A. C., Velazquez, F. R., Perez-Schael, I., Saez-Llorens, X., Abate, H., Espinoza, F.,

Lopez, P., Macias-Parra, M., Ortega-Barria, E., Rivera-Medina, D. M., Rivera, L., Pavia-

Ruz, N., Nunez, E., Damaso, S., Ruiz-Palacios, G. M., De Vos, B., O'ryan, M., Gillard, P.

& Bouckenooghe, A. 2008. Efficacy and safety of an oral live attenuated human rotavirus

vaccine against rotavirus gastroenteritis during the first 2 years of life in Latin American

infants: a randomised, double-blind, placebo-controlled phase III study. Lancet, 371, 1181-

9.

54

Liu, L., Oza, S., Hogan, D., Perin, J., Rudan, I., Lawn, J. E., Cousens, S., Mathers, C. & Black, R.

E. 2015a. Global, regional, and national causes of child mortality in 2000-13, with

projections to inform post-2015 priorities: an updated systematic analysis. Lancet, 385,

430-40.

Liu, L., Oza, S., Hogan, D., Perin, J., Rudan, I., Lawn, J. E., Cousens, S., Mathers, C. & Black, R.

E. 2015b. Global, regional, and national causes of child mortality in 2000–13, with

projections to inform post-2015 priorities: an updated systematic analysis. The Lancet, 385,

430-440.

Luchs, A., Cilli, A., Morillo, S. G., De Cassia Compagnoli Carmona, R. & Do Carmo Sampaio

Tavares Timenetsky, M. 2014. Rotavirus in adults, Brazil, 2004-2011: G2P[4] dominance

and potential impact on vaccination. Brazilian Journal of Infectious Diseases, 18, 53-9.

Lynch, M., Shieh, W. J., Bresee, J. S., Tatti, K. M., Gentsch, J. R., Jones, T., Jiang, B.,

Hummelman, E., Zimmerman, C. M., Zaki, S. R. & Glass, R. I. 2006. Intussusception after

administration of the rhesus tetravalent rotavirus vaccine (Rotashield): the search for a

pathogenic mechanism. Pediatrics, 117, e827-32.

Madhi, S. A., Cunliffe, N. A., Steele, D., Witte, D., Kirsten, M., Louw, C., Ngwira, B., Victor, J.

C., Gillard, P. H., Cheuvart, B. B., Han, H. H. & Neuzil, K. M. 2010. Effect of Human

Rotavirus Vaccine on Severe Diarrhea in African Infants. New England Journal of

Medicine, 362, 289-298.

Maes, P., Matthijnssens, J., Rahman, M. & Van Ranst, M. 2009. RotaC: a web-based tool for the

complete genome classification of group A rotaviruses. BMC Microbiology, 9, 238.

Mandeville, K. L., Krabshuis, J., Ladep, N. G., Mulder, C. J. J., Quigley, E. M. M. & Khan, S. A.

2009. Gastroenterology in developing countries: issues and advances. World journal of

gastroenterology, 15, 2839-2854.

Marcelin, G., Miller, A. D., Blutt, S. E. & Conner, M. E. 2011. Immune mediators of rotavirus

antigenemia clearance in mice. Journasl of Virology, 85, 7937-41.

Markkula, J., Hemming-Harlo, M. & Vesikari, T. 2014. Detection of Vaccine-Derived Rotavirus

Strains in Non-Immunocompromised Children up to 3-6 Months after RotaTeq®

Vaccination.

Martorell, R., Horta, B., Adair, L., Stein, A., Richter, L., H.D. Fall, C., Bhargava, S., Dey Biswas,

S. K., Perez, L., C Barros, F. & G Victora, C. 2009. Weight Gain in the First Two Years of

Life Is an Important Predictor of Schooling Outcomes in Pooled Analyses from Five Birth

Cohorts from Low- and Middle-Income Countries.

Masibo, P. K. & Makoka, D. 2012. Trends and determinants of undernutrition among young

Kenyan children: Kenya Demographic and Health Survey; 1993, 1998, 2003 and 2008-

2009. Public Health and Nutrition, 15, 1715-27.

55

Matthijnssens, J., Ciarlet, M., Heiman, E., Arijs, I., Delbeke, T., Mcdonald, S. M., Palombo, E. A.,

Iturriza-Gomara, M., Maes, P., Patton, J. T., Rahman, M. & Van Ranst, M. 2008. Full

genome-based classification of rotaviruses reveals a common origin between human Wa-

Like and porcine rotavirus strains and human DS-1-like and bovine rotavirus strains.

Journal of Virology, 82, 3204-19.

Matthijnssens, J., Ciarlet, M., Mcdonald, S. M., Attoui, H., Banyai, K., Brister, J. R., Buesa, J.,

Esona, M. D., Estes, M. K., Gentsch, J. R., Iturriza-Gomara, M., Johne, R., Kirkwood, C.

D., Martella, V., Mertens, P. P., Nakagomi, O., Parreno, V., Rahman, M., Ruggeri, F. M.,

Saif, L. J., Santos, N., Steyer, A., Taniguchi, K., Patton, J. T., Desselberger, U. & Van

Ranst, M. 2011a. Uniformity of rotavirus strain nomenclature proposed by the Rotavirus

Classification Working Group (RCWG). Archives of Virology, 156, 1397-413.

Matthijnssens, J., De Grazia, S., Piessens, J., Heylen, E., Zeller, M., Giammanco, G. M., Banyai,

K., Buonavoglia, C., Ciarlet, M., Martella, V. & Van Ranst, M. 2011b. Multiple

reassortment and interspecies transmission events contribute to the diversity of feline,

canine and feline/canine- like human group A rotavirus strains. Infections and Genetic

Evolutions, 11, 1396-406.

Matthijnssens, J., Otto, P. H., Ciarlet, M., Desselberger, U., Van Ranst, M. & Johne, R. 2012. VP6-sequence-based cutoff values as a criterion for rotavirus species demarcation. Archives of Virology, 157, 1177-82.

Matthijnssens, J., Zeller, M., Heylen, E., De Coster, S., Vercauteren, J., Braeckman, T., Van Herck,

K., Meyer, N., Pircon, J. Y., Soriano-Gabarro, M., Azou, M., Capiau, H., De Koster, J.,

Maernoudt, A. S., Raes, M., Verdonck, L., Verghote, M., Vergison, A., Van Damme, P. &

Van Ranst, M. 2014. Higher proportion of G2P[4] rotaviruses in vaccinated hospitalized

cases compared with unvaccinated hospitalized cases, despite high vaccine effectiveness

against heterotypic G2P[4] rotaviruses. Clinical Microbiology and Infections, 20, O702-

10.

Mcdonald, S. M. & Patton, J. T. 2008. Molecular characterization of a subgroup specificity

associated with the rotavirus inner capsid protein VP2. Journal of virology, 82, 2752-2764.

Mcdonald, S. M. & Patton, J. T. 2011. Rotavirus VP2 Core Shell Regions Critical for Viral

Polymerase Activation. Journal of Virology, 85, 3095-3105.

Meshram, I., Arlappa, N., Balakrishna, N., Laxmaiah, A., Kodavanti, M., Chitty, G. R.,

Ravindranath, M., Sharad Kumar, S. & Brahmam, G. 2011. Prevalence and Determinants

of Undernutrition and its Trends among Pre-School Tribal Children of Maharashtra State,

India.

Mondal, D., Haque, R., Sack, R. B., Kirkpatrick, B. D. & Petri, W. A., Jr. 2009. Attribution of

malnutrition to cause-specific diarrheal illness: evidence from a prospective study of

preschool children in Mirpur, Dhaka, Bangladesh. American Journal of Tropical Medicine

and Hygiene, 80, 824-6.

Moon, S. S., Wang, Y., Shane, A. L., Nguyen, T., Ray, P., Dennehy, P., Baek, L. J., Parashar, U.,

Glass, R. I. & Jiang, B. 2010. Inhibitory effect of breast milk on infectivity of live oral

rotavirus vaccines. Pediatric Infectious Diseases Journal, 29, 919-23.

56

Moore, S. E., Goldblatt, D., Bates, C. J. & Prentice, A. M. 2003. Impact of nutritional status on

antibody responses to different vaccines in undernourished Gambian children. Acta

Paediatrica, 92, 170-6.

Mora, J. R., Iwata, M. & Von Andrian, U. H. 2008. Vitamin effects on the immune system:

vitamins A and D take centre stage. Nature Reviews Immunology, 8, 685-98.

Mpabalwani, E. M., Simwaka, C. J., Mwenda, J. M., Mubanga, C. P., Monze, M., Matapo, B.,

Parashar, U. D. & Tate, J. E. 2016. Impact of Rotavirus Vaccination on Diarrheal

Hospitalizations in Children Aged <5 Years in Lusaka, Zambia. Clinical Infectious

Diseases, 62 Suppl 2, S183-7.

Mpabalwani, M., Oshitani, H., Kasolo, F., Mizuta, K., Luo, N., Matsubayashi, N., Bhat, G.,

Suzuki, H. & Numazaki, Y. 1995. Rotavirus gastro-enteritis in hospitalized children with

acute diarrhoea in Zambia. Annals of tropical paediatrics, 15, 39-43.

Mukhopadhya, I., Sarkar, R., Menon, V., Babji, S., Paul, A., Rajendran, P., Thuppal, S., Moses,

P., Iturriza-Gòmara, M., Gray, J. & Kang, G. 2013. Rotavirus Shedding in Symptomatic

and Asymptomatic Children using Reverse Transcription-Quantitative PCR.

Myatt, M., Khara, T. & Collins, S. 2006. A review of methods to detect cases of severely

malnourished children in the community for their admission into community-based

therapeutic care programs. Food and Nutrition Bulletin, 27, S7-23.

Nelson, E. A. & Glass, R. I. 2010. Rotavirus: realising the potential of a promising vaccine.

Lancet, 376, 568-70.

Olofin, I., Mcdonald, C. M., Ezzati, M., Flaxman, S., Black, R. E., Fawzi, W. W., Caulfield, L. E.

& Danaei, G. 2013. Associations of suboptimal growth with all-cause and cause-specific

mortality in children under five years: a pooled analysis of ten prospective studies. PLoS

One, 8, e64636.

Organization, W. H. 2009. Global prevalence of vitamin A deficiency in populations at risk 1995-

2005: WHO global database on vitamin A deficiency.

Organization, W. H. 2016. World health statistics 2016: monitoring health for the SDGs

sustainable development goals, World Health Organization.

Page, A.-L., De Rekeneire, N., Sayadi, S., Aberrane, S., Janssens, A.-C., Rieux, C., Djibo, A.,

Manuguerra, J.-C., Ducou-Le -Pointe, H., Grais, R. F., Schaefer, M., Guerin, P. J. & Baron,

E. 2013. Infections in children admitted with complicated severe acute malnutrition in

Niger. PloS one, 8, e68699-e68699.

Parashar, U. D., Gibson, C. J., Bresee, J. S. & Glass, R. I. 2006. Rotavirus and severe childhood

diarrhea. Emerging infectious diseases, 12, 304-306.

Parra, M., Herrera, D., Calvo-Calle, J. M., Stern, L. J., Parra-López, C. A., Butcher, E., Franco,

M. & Angel, J. 2014. Circulating human rotavirus specific CD4 T cells identified with a

class II tetramer express the intestinal homing receptors α4β7 and CCR9. Virology, 452-

453, 191-201.

57

Pasetti, M. F., Simon, J. K., Sztein, M. B. & Levine, M. M. 2011. Immunology of gut mucosal

vaccines. Immunology Review, 239, 125-48.

Payne, D. C., Boom, J. A., Staat, M. A., Edwards, K. M., Szilagyi, P. G., Klein, E. J., Selvarangan,

R., Azimi, P. H., Harrison, C., Moffatt, M., Johnston, S. H., Sahni, L. C., Baker, C. J.,

Rench, M. A., Donauer, S., Mcneal, M., Chappell, J., Weinberg, G. A., Tasslimi, A., Tate,

J. E., Wikswo, M., Curns, A. T., Sulemana, I., Mijatovic-Rustempasic, S., Esona, M. D.,

Bowen, M. D., Gentsch, J. R. & Parashar, U. D. 2013. Effectiveness of pentavalent and

monovalent rotavirus vaccines in concurrent use among US children <5 years of age, 2009-

2011. Clinical Infectious Diseases, 57, 13-20.

Petri, W. A., Jr., Miller, M., Binder, H. J., Levine, M. M., Dillingham, R. & Guerrant, R. L. 2008.

Enteric infections, diarrhea, and their impact on function and development. Journal of

Clinical Investigations, 118, 1277-90.

Pino-Lagos, K., Benson, M. J. & Noelle, R. J. 2008. Retinoic acid in the immune system. Annals

of the New York Academy of Sciences, 1143, 170-87.

Pott, J., Mahlakoiv, T., Mordstein, M., Duerr, C. U., Michiels, T., Stockinger, S., Staeheli, P. &

Hornef, M. W. 2011. IFN-lambda determines the intestinal epithelial antiviral host defense.

Proceedings of the National Academy of Sciences of the United States of America, 108,

7944-9.

Prendergast, A. J. 2015. Malnutrition and vaccination in developing countries. Philosophical

transactions of the Royal Society of London. Series B, Biological sciences, 370, 20140141.

Qadri, F., Wierzba, T. F., Ali, M., Chowdhury, F., Khan, A. I., Saha, A., Khan, I. A., Asaduzzaman,

M., Akter, A., Khan, A., Begum, Y. A., Bhuiyan, T. R., Khanam, F., Chowdhury, M. I.,

Islam, T., Chowdhury, A. I., Rahman, A., Siddique, S. A., You, Y. A., Kim, D. R., Siddik,

A. U., Saha, N. C., Kabir, A., Cravioto, A., Desai, S. N., Singh, A. P.

& Clemens, J. D. 2016. Efficacy of a Single-Dose, Inactivated Oral Cholera Vaccine in

Bangladesh. New England Journal of Medicine, 374, 1723-32.

Ray, P. G., Kelkar, S. D., Walimbe, A. M., Biniwale, V. & Mehendale, S. 2007. Rotavirus

immunoglobulin levels among Indian mothers of two socio-economic groups and

occurrence of rotavirus infections among their infants up to six months. Journal of Medical

Virology, 79, 341-349.

Rha, B., Tate, J. E., Weintraub, E., Haber, P., Yen, C., Patel, M., Cortese, M. M., Destefano, F.

& Parashar, U. D. 2014. Intussusception following rotavirus vaccination: an updated

review of the available evidence. Expert Review on Vaccines, 13, 1339-48.

Richardson, V., Hernandez-Pichardo, J., Quintanar-Solares, M., Esparza-Aguilar, M., Johnson, B.,

Gomez-Altamirano, C. M., Parashar, U. & Patel, M. 2010. Effect of rotavirus vaccination

on death from childhood diarrhea in Mexico. The New England journal of medicine, 362,

299-305.

58

Rodriguez -Limas, W. A., Pastor, A. R., Esquivel-Soto, E., Esquivel-Guadarrama, F., Ramirez, O.

T. & Palomares, L. A. 2014. Immunogenicity and protective efficacy of yeast extracts

containing rotavirus-like particles: a potential veterinary vaccine. Vaccine, 32, 2794-8.

Rosillon, D., Buyse, H., Friedland, L. R., Ng, S. P., Velazquez, F. R. & Breuer, T. 2015. Risk of

Intussusception After Rotavirus Vaccination: Meta-analysis of Postlicensure Studies.

Pediatic Infectious Diseases Journal, 34, 763-8.

Ruiz-Palacios, G. M., Perez-Schael, I., Velazquez, F. R., Abate, H., Breuer, T., Clemens, S. C.,

Cheuvart, B., Espinoza, F., Gillard, P., Innis, B. L., Cervantes, Y., Linhares, A. C., Lopez,

P., Macias-Parra, M., Ortega-Barria, E., Richardson, V., Rivera-Medina, D. M., Rivera, L.,

Salinas, B., Pavia- Ruz, N., Salmeron, J., Ruttimann, R., Tinoco, J. C., Rubio, P., Nunez,

E., Guerrero, M. L., Yarzabal, J. P., Damaso, S., Tornieporth, N., Saez-Llorens, X.,

Vergara, R. F., Vesikari, T., Bouckenooghe, A., Clemens, R., De Vos, B. & O'ryan, M.

2006. Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis.

New England Journal of Medicine, 354, 11-22.

Rytter, M., Kolte, L., Briend, A., Friis, H. & Brix Christensen, V. 2014. The Immune System in

Children with Malnutrition-A Systematic Review.

Sanders, D., Yukich, J., Shrimpton, R., Greiner, T. & Mason, J. 2014. Vitamin A policies need

rethinking. International Journal of Epidemiology, 44, 283-292.

Santos, N. & Hoshino, Y. 2005. Global distribution of rotavirus serotypes/genotypes and its

implication for the development and implementation of an effective rotavirus vaccine.

Review of Medical Virology, 15, 29-56.

Sapparapu, G., Sims, A. L., Aiyegbo, M. S., Shaikh, F. Y., Harth, E. M. & Crowe, J. E., Jr. 2014. Intracellular neutralization of a virus using a cell-penetrating molecular transporter. Nanomedicine (London), 9, 1613-24.

Savy, M., Edmond, K., Fine, P. E., Hall, A., Hennig, B. J., Moore, S. E., Mulholland, K., Schaible,

U. & Prentice, A. M. 2009. Landscape analysis of interactions between nutrition and

vaccine responses in children. Journal of Nutrition, 139, 2154s-218s.

Sen, A., Pruijssers, A. J., Dermody, T. S., Garcia-Sastre, A. & Greenberg, H. B. 2011. The early

interferon response to rotavirus is regulated by PKR and depends on MAVS/IPS-1, RIG-I,

MDA-5, and IRF3. Journal of Virology, 85, 3717-32.

Settembre, E. C., Chen, J. Z., Dormitzer, P. R., Grigorieff, N. & Harrison, S. C. 2011. Atomic

model of an infectious rotavirus particle. Emboryology Journal, 30, 408-16.

Simonsen, L., Viboud, C., Elixhauser, A., Taylor, R. J. & Kapikian, A. Z. 2005. More on

RotaShield and intussusception: the role of age at the time of vaccination. Journal of

Infectious Diseases, 192 Suppl 1, S36-43.

59

Tate, J. E., Burton, A. H., Boschi-Pinto, C., Steele, A. D., Duque, J. & Parashar, U. D. 2012. 2008

estimate of worldwide rotavirus-associated mortality in children younger than 5 years

before the introduction of universal rotavirus vaccination programmes: a systematic review

and meta-analysis. The Lancet Infectious Diseases, 12, 136-141.

Tate, J. E., Ngabo, F., Donnen, P., Gatera, M., Uwimana, J., Rugambwa, C., Mwenda, J. M. &

Parashar, U. D. 2016. Effectiveness of Pentavalent Rotavirus Vaccine Under Conditions

of Routine Use in Rwanda. Clinical Infections and Diseases, 62 Suppl 2, S208-12.

Todd, S., Page, N. A., Duncan Steele, A., Peenze, I. & Cunliffe, N. A. 2010. Rotavirus strain types

circulating in Africa: Review of studies published during 1997-2006. Journal of Infectious

Diseases, 202 Suppl, S34-42.

Trask, S. D., Kim, I. S., Harrison, S. C. & Dormitzer, P. R. 2010. A rotavirus spike protein

conformational intermediate binds lipid bilayers. Journal of Virology, 84, 1764-70.

Uauy, R., Kain, J. & Corvalan, C. 2011. How can the Developmental Origins of Health and Disease

(DOHaD) hypothesis contribute to improving health in developing countries? American

Journal of Clinical Nutrition, 94, 1759s-1764s.

Verkerke, H., Sobuz, M. S., Z. Ma, J., E. Petri, S., Reichman, D., Qadri, F., Rahman, M., Haque,

R. & A. Petri, W. 2016. Malnutrition Is Associated with Protection from Rotavirus

Diarrhea: Evidence from a Longitudinal Birth Cohort Study in Bangladesh.

Vesikari, T., Karvonen, A., Prymula, R., Schuster, V., Tejedor, J. C., Cohen, R., Meurice, F., Han,

H. H., Damaso, S. & Bouckenooghe, A. 2007. Efficacy of human rotavirus vaccine against

rotavirus gastroenteritis during the first 2 years of life in European infants: randomised,

double-blind controlled study. Lancet, 370, 1757-63.

Vesikari, T., Matson, D. O., Dennehy, P., Van Damme, P., Santosham, M., Rodriguez, Z., Dallas,

M. J., Heyse, J. F., Goveia, M. G., Black, S. B., Shinefield, H. R., Christie, C. D., Ylitalo,

S., Itzler, R. F., Coia, M. L., Onorato, M. T., Adeyi, B. A., Marshall, G. S., Gothefors, L.,

Campens, D., Karvonen, A., Watt, J. P., O'brien, K. L., Dinubile, M. J., Clark, H. F.,

Boslego, J. W., Offit, P. A. & Heaton, P. M. 2006. Safety and efficacy of a pentavalent

human-bovine (WC3) reassortant rotavirus vaccine. New England Journal of Medicine,

354, 23-33.

Vesikari, T., Uhari, M., Renko, M., Hemming, M., Salminen, M., Torcel-Pagnon, L., Bricout, H.

& Simondon, F. 2013. Impact and effectiveness of RotaTeq(R) vaccine based on 3 years

of surveillance following introduction of a rotavirus immunization program in Finland.

Pediatric Infectious Diseases Journal, 32, 1365-73.

Von Gottberg, A., De Gouveia, L., Tempia, S., Quan, V., Meiring, S., Von Mollendorf, C., Madhi,

S. A., Zell, E. R., Verani, J. R., O’brien, K. L., Whitney, C. G., Klugman, K. P. & Cohen,

C. 2014. Effects of Vaccination on Invasive Pneumococcal Disease in South Africa. New

England Journal of Medicine, 371, 1889-1899.

60

Walker, C. L. F., Rudan, I., Liu, L., Nair, H., Theodoratou, E., Bhutta, Z. A., O'brien, K. L.,

Campbell, H. & Black, R. E. 2013. Global burden of childhood pneumonia and diarrhoea.

Lancet, 381, 1405-1416.

Weitkamp, J. H., Kallewaard, N. L., Bowen, A. L., Lafleur, B. J., Greenberg, H. B. & Crowe, J.

E., Jr. 2005. VH1-46 is the dominant immunoglobulin heavy chain gene segment in

rotavirus-specific memory B cells expressing the intestinal homing receptor alpha4beta7.

ournal of Immunology, 174, 3454-60.

Who, M. 2006. Growth. Reference, Study, Group: WHO Child Growth Standards: Length/height-

for-age, weight-for-age, weight -for-length, weight-for-height and body mass index-for-

age: Methods and development.

Wolf, M., Vo, P. T. & Greenberg, H. B. 2011. Rhesus Rotavirus Entry into a Polarized Epithelium

Is Endocytosis Dependent and Involves Sequential VP4 Conformational Changes. Journal

of Virology, 85, 2492-2503.

Yuan, L., Wen, K., Azevedo, M. S. P., Gonzalez, A. M., Zhang, W. & Saif, L. J. 2008. Virus-

specific intestinal IFN-gamma producing T cell responses induced by human rotavirus

infection and vaccines are correlated with protection against rotavirus diarrhea in

gnotobiotic pigs. Vaccine, 26, 3322-3331.

Zandoh, C., Adjei, G., Tchum, S., Newton, S., Owusu-Agyei, S., Kirkwood, B. R., Filteau, S.,

Ampofo, W. & Adjuik, M. 2007. Vitamin A Supplementation Enhances Infants' Immune Responses to Hepatitis B Vaccine but Does Not Affect Responses to Haemophilus

influenzae Type b Vaccine. The Journal of Nutrition, 137, 1272-1277.

Zeng, R., Oderup, C., Yuan, R., Lee, M., Habtezion, A., Hadeiba, H. & Butcher, E. C. 2013.

Retinoic acid regulates the development of a gut-homing precursor for intestinal dendritic

cells. Mucosal Immunology, 6, 847-56.

61

APPENDICES

Appendix A: Letters of support.

62

63

64

Appendix B: Study questionnaire and clinical examination form.

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

CASE RECORD FORM

ID NUMBER:

CAUSES OF ROTAVIRUS VACCINE FAILURE IN ZAMBIA ROVAS STUDY

PART A –VISIT 1.0

ELIGIBILITY CHECK

65

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

INCLUSION CRITERIA (All must be yes) YES NO

1. Mother willing to participate voluntarily and able to provide signed informed

consent (with witness in the case of illiterate participant)

2. Infant eligible for rotavirus vaccine immunization as per national policy (male or female infant, 6-12 weeks old)

3. Mother willing to undergo study procedures, including questionnaires, HIV counselling and

testing, CD4 testing, and provide breast milk and blood sample at enrolment.

4. Mother willing for child to undergo study procedures including full-course rotavirus vaccination, phlebotomy at enrolment and 1 month post-rotavirus vaccination, quarterly visits

and presentation to clinic for collection of stool sample when infant has diarrhoea.

5. Plans to remain resident in the area and willing to come for scheduled visits for the

duration of the study.

EXCLUSION CITERIA (All must be No) YES NO

1. Child has contraindication to rotavirus vaccination.

2. Previous administration of rotavirus vaccine to child.

3. Recent immunosuppressive therapy in child (including high-dose systemic corticosteroids).

4. History of ever receiving a blood transfusion or blood products, including immunoglobulins within the last 6 months, for mother and child. 5. Any condition deemed by the study investigator to pose potential harm to the participants or

jeopardize the validity of study results.

AGE OF MOTHER__________________ (In years)

INFANT AGE _____________ INFANT SEX (In weeks) (M/F)

PART B – VISIT 1.0 BASELINE PROFILE

66

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

DATE OFBASESLINE VISIT________/______/_________ (dd mm yyyyy).KEY MEDICAL HISTORY

DATE OF BIRTH ________/______/__________

(ddmmyyyy) PLACE OF BIRTH(Hospital=1, clinic=2, Health post=3, Home=4, Other=5) HOSPITALIZATION SINCE BIRTH?(State Yes or No; and if Yes, dates of hospitalization)

1. ________/______/__________ (dd mm yyyy)

2. ________/______/__________

3. (dd mm yyyy) ________/______/__________

(dd mm yyyy)

IMMUNIZATION/SUPPLEMENTS RECORD

(Indicate date administered)

KNOWN DISEASE THAT WOULD AFFECT

IMMUNOLOGICAL RESPONSE TO VACCINE? BCG OPV0 PENT1 VIT A ALBENDAZOL

(Stage 4 AIDS= 1, Lymphoma=2, other DATE Malignancies=3, Steroid treatment=4,

Other=5

[PLEASE DO NOT IMMUNISE IF ANY OF THESE ARE PRESENT]

MATERNAL HIV STATUS

(Positive=1; Negative=2).

MATERNAL CD4 IF HIV POSITIVE (Not Applicable HIV –ve) ___________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy,

specify)______________________________________________________________________________________ ___

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

LABORATORY SAMPLES Have samples for the following been collected? If yes, tick appropriate boxand arrange results

follow up MATERNAL BREAST MILK IgA AND IgG ___________________

INFANT BLOOD FOR ZINC& VITAMIN A

____________________

INFANT BLOOD FOR IMMUNOGENICITYS ___________________

ESCORT THE MOTHER/INFANT FOR ROUTINE IMMUNIZATION (INCLUDING ROTAVIRUS) AND PROVIDE INFORMATION FOR THE NEXT

EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL.

67

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA ROTAVIRUS VACCINE DOSE 1 BATCH NUMBER: _______________________________________ NEXT VISIT DATE____/____/_______

VISIT 2.0

FOLLOW UP

DATE OF VISIT________/______/__________ (dd mm yyyyy)

DATE OF RV1 VACCINATION ________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED ______________________________________________________________

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

ESCORT THE MOTHER/INFANT FOR ROUTINE IMMUNIZATION (INCLUDING ROTAVIRUS) AND PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL.

ROTAVIRUS VACCINE DOSE 2 LOT NUMBER: __________________________________________ NEXT VISIT DATE____/____/_______

68

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

VISIT 3.0

FOLLOW UP

DATE OF VISIT________/______/__________ (dd mm yyyyy)

DATE OF RV2 VACCINATION ________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

LABORATORY SAMPLES Have samples for the following been collected? If yes plan to follow up on results

INFANT BLOOD FOR IMMUNOGENICITY ______________________________________ (Enter results when

collected)_______________________________________

69

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL NEXT VISIT DATE____/____/_______

VISIT 4.0

FOLLOW UP DATE OF VISIT________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

70

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL. NEXT VISIT DATE____/____/_______

VISIT 5.0

FOLLOW UP

DATE OF VISIT________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION

GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO

IN CASE INFANT IS UNWELL. NEXT VISIT DATE____/____/_______

71

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

VISIT 6.0

FOLLOW UP

DATE OF VISIT________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION

GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

LABORATORY SAMPLES

INFANT BLOOD FOR IMMUNIGENICITY ______________________________________ (Enter results when

collected)_______________________________________

72

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL. NEXT VISIT DATE____/____/_______

VISIT

QUARTERLY DURING MONTHS 15-42

DATE OF VISIT________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIARRHOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

PROVIDE INFORMATION FOR THE NEXT EXPECTED VISIT AND INSTRUCTION ON WHAT TO DO IN CASE INFANT IS UNWELL. NEXT VISIT DATE____/____/_______

73

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

VISIT

CLOSE OUT VISIT AT MONTH 42

DATE OF VISIT________/______/__________ (dd mm yyyyy)

KEY MEDICAL HISTORY

HAS THE INFANT BEEN UNWELL SINCE THE LAST VISIT?

WAS THE INFANT TAKEN TO A HEALTH FACILITY?

WHAT MEDICATION WAS ADMINISTERED

DID THE INFANT HAVE ANY DIRAHOOEAL EPISODE SINCE THE LAST VISIT? (If yes, provide date and ensure that the unscheduled sick visit form is completed)

WAS STOOL SAMPLE PROVIDED? (If yes, enter the laboratory results)

_____________________________________ _____________________________________ _____________________________________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy, specify)

__________________________________________________________________________________________

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

LABORATORY SAMPLES

INFANT BLOOD FOR IMMUNIGENICITY ______________________________________ (Enter results when

collected)_______________________________________

74

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA THANK THE MOTHER FOR HAVING PARTICIPATED IN THE STUDY AND PROVIDE A DATE

WHEN FINAL RESULTS FROM THE STUDY WILL BE COMMUNICATED.

VISIT UNSCHEDULED “SICK” VISIT FORM

DATE OF VISIT________/______/__________

(dd mm yyyyy)

KEY MEDICAL HISTORY WHAT IS THE MAIN PROBLEM FOR THIS VISIT? _______________________________________________________________________________________________________________ _______________________________________________________________________________________________________________ _______________________________________________________________________________________________________________

Did the child have any diarrhoea since last visit?

if yes, date diarrhoea started ____/_____/________ if yes, date diarrhoea ended____/____/______

Did child have 3 or more loose or watery stools in the last 24 hours? Was stool sample collected during diarrhoea bout? What day was the stool sample collected______/______/________

GENERAL MEDICAL EXAMINATION GENERAL MEDICAL CONDITION OF INFANT (Healthy=1; Not healthy=2; If not healthy,

specify)______________________________________________________________________________________ ____

WEIGHT (in Kilogrammes)

HEIGHT(in centimetres)

MUAC(Mid Upper Arm Circumference in centimetres)

SKIN FOLD (in mm) TRICEPS____________________SUBSCAPULAR_________________________

LABORATORY SAMPLES

Has any laboratory sample been collected? If yes, specify _________________________________ _________________________________ _________________________________

SEVERE GASTROENTERISTIS ASSESSMENT SEVERITY OF CURRENT GE EPISODE (No dehydration=1, Some Dehydration=2, Severe Dehydration=3)

VESIKARI SCORE (Use study specific Vesikari score form)

75

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

IF ADMITED TO HOSPITAL DURATION OF HOSPITAL ADMISION

FINAL DIAGNOSIS_______________________________________________________________________

Adverse Experience Points

Duration of looser than normal stools (days) 1

1- 4 2 5

3

≥ 6

____/____/_______

MODI FIED VESIK ARI SCALE

EVALUATI

NG AND SCORING SEVERITY OF GASTROE NTERISTIS

STUDY ID NUMBER DATE EVALUAT ED:

76

CENTRE FOR INFECTIOUS DISEASE RESEARCH IN ZAMBIA

Maximum # of looser than normal stools per 24 hours 1 1-3 2

4-5 3

≥ 6

Duration of vomiting (days) 1 1 2 2 3

≥ 3

Maximum # of episodes of vomiting per 24 hours 1 1 2

2-4 3

≥ 5

Fever (highest temperature recorded during episode)

Rectally Axillary 1 37.1 – 38.4°C 36.6 – 37.9°C 2

38.5 – 38.9°C 38.0 – 38.4°C 3

≥ 39°C ≥ 38.5°C

Dehydration 1-5% 2

≥ 6% 3 TOTAL

Treatment

SCORE: Rehydration 1 _____________

Hospitalization 2 ______


Recommended