vaccination strategy to protect against flavivirus ...€¦ · solution to complement the current...

135
Vaccination Strategy To Protect Against Flavivirus Infection Based On Recombinant Measles Vaccine by Rowida Abdelgalel A Dissertation Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Approved November 2016 by the Graduate Supervisory Committee: Jorge Reyes del Valle, Co-Chair Hugh Mason, Co-Chair Douglas Lake Valerie Stout Wayne Frasch ARIZONA STATE UNIVERSITY December 2016

Upload: others

Post on 11-Jun-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

Vaccination Strategy To Protect Against Flavivirus Infection Based On Recombinant

Measles Vaccine

by

Rowida Abdelgalel

A Dissertation Presented in Partial Fulfillment of the Requirements for the Degree

Doctor of Philosophy

Approved November 2016 by the Graduate Supervisory Committee:

Jorge Reyes del Valle, Co-Chair

Hugh Mason, Co-Chair Douglas Lake Valerie Stout Wayne Frasch

ARIZONA STATE UNIVERSITY

December 2016

Page 2: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

i

ABSTRACT

Despite the approval of a Dengue virus (DV) vaccine in five endemic countries,

dengue prevention would benefit from an immunization strategy highly immunogenic in

young infants and not curtailed by viral interference. Problematically, infants younger

than 9 year of age, whom are particularly prone to Dengue severe infection and death,

cannot be immunized using current approved DV vaccine. The most important issues

documented so far are the lack of efficiency and enhancement of the disease in young

seronegative recipients, as well as uneven protection against the four DV serotypes.

Based on data from clinical trials that showed enhanced performance of dengue vaccines

when the host has previous anti-flaviviral immunity, I proposed here an attractive

solution to complement the current vaccine: a recombinant measles vaccine vectoring

dengue protective antigens to be administered to young infants. I hypothesized that

recombinant measles virus expressing Dengue 2 and 4 antigens would successfully

induce neutralizing responses against DV2 and 4 and the vaccine cocktail of this

recombinant measles can prime anti-flaviviral neutralizing immunity. For this

dissertation, I generated and performed preclinical immune assessment for four novel

Measles-Dengue (MV-DV) vaccine candidates. I generated four MVs expressing the pre

membrane (prM) and full length or truncated (90%) forms of the major envelope (E)

from DV2 and DV4. Two virus, MVvac2-DV2(prME)N and MVvac2-DV4(prME),

expressed high levels of membrane associated full-length E, while the other two viruses,

MVvac2-DV2(prMEsol)N and MVvac2-DV4(prMEsol)N, expressed and secreted

truncated, soluble E protein to its extracellular environment. The last two vectored

Page 3: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

ii

vaccines proved superior anti-dengue neutralizing responses comparing to its

corresponding full length vectors. Remarkably, when MVvac2-DV2/4(prMEsol)N

recombinant vaccines were combined, the vaccine cocktail was able to prime cross-

neutralizing responses against DV 1 and the relatively distant 17D yellow fever virus

attenuated strain. Thus, I identify a promising DV vaccination strategy, MVvac2-

DV2/4(prMEsol)N, which can prime broad neutralizing immune responses by using only

two of the four available DV serotypes. The current MV immunization scheme can be

advantageus to prime broad anti-flaviviral neutralizing immunity status, which will be

majorly boosted by subsequent chimeric Dengue vaccine approaches.

Page 4: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

iii

Didicated to the joy of my life, my son: Saleem Mousa

Page 5: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

iv

ACKNOWLEDGMENTS

Foremost, I would like to express my sincere gratitude to my mintor Dr. Jorge

Reyes del Valle for his help during this journey. The work that I have done during my

PhD was mainly done because of his guidance and support. He showed a great deal of

confidence in my abilities and he belives in me. He has been a professional model for me

and a great lifelong firend. I also want to thank my committee members, Dr. Hugh

Mason, Dr. Douglas Lake, Dr. Valerie Stout, and Dr. Wayne Frasch, for their support and

feedback. It is because of all of them that I was able to grow as a scientist, and to learn to

challenge my knowledge and expand my limits.

During my time as a PhD student I was lucky enough to share my experience with

special friends, Ivonne, Emily, Indera, Francisca and Radwa. They gave me great support

and helped me to get over the hard times, esspacially, Ivonne gave me great technical

support.

Last but not the least, I would like to thank a dear friend Wijdan Al Omari for her

help, she gave me lots of support and helped me through the hard times, she is not just a

friend she is my sister as well. I would like to thanks my family members, my joy of life,

my son, Saleem and my husband Mohsen, my parents Zainab and Abdelhameed who

belived on me and gave me love and support.

Page 6: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

v

TABLE OF CONTENTS

Page

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

CHAPTER

1 INTRODUCTION…………………………….…………………………………..1

The Measles Virus Reverses Genetics System In The Development Of

Multivalent Vaccine Options.…………………...………………………...…….1

The Measles Virus Biology…….…………………………………………1

The Rescue Of Measles Virus From Cloned DNA..……………………...3

Genomic Modification Of MV, The Insertion Of Foreign Coding

Sequence…………………………………………………………………..4

Measles Candidate Multivalent Recombinant Vaccines…………………..5

The Quest For An Optimal, Pediatric Dengue Virus Vaccine.…………………..7

Epidemiology Of Dengue Virus (DV)…………………………………….7

The Pathogenesis Of Dengue Infection...…………………………………9

Dengue Virus Biology……………………………………………...……10

Immune Response And Immunopathogenesis Of Dengue Virus

Infection.…………………………………………………………………13

Dengue Vaccine Candidates.…………………………………………….16

Central Hypothesis.…………………………………………………………….20

Chapter Organization…………………………………………………………..21

References……………………………………………………………………..25

Page 7: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

vi

CHAPTER Page

2 BROAD NEUTRALIZING RESPONSES ELICITED BY FLAVIVIRUS

INFECTION IN CD46 INFarKO MICE……….…….………………………….32

Abstract………………………………………………………………………32

Importance……………………………………………………………………33

Introduction…………………………………………………………………..34

Materials And Methods………………………………………………………36

Cells And Viruses………………………………………………………..36

Mice Inoculations………………………………………………….……..36

DV And YfV Neutralization Assay……………………………….……..36

Correlation Of PRNT50 And LNI…………………………………….…37

Results…………………………………………………………………….…..38

Monotypic Dengue Virus Immunity………………………………….….38

Heterologous Cross-Neutralization Immunity To DV1 And YFV………39

Discussion…………………………………………………………………….40

References……………………………………………………………………..44

3 GENERATION OF A COMPLEMENTARY ANTI-DENGUE

IMMUNIZATION STRATEGY BASED ON A RECOMBINANT MEASLES

VIRUS VACCINE COCKTAIL……………………………..……………………….46

Abstract………………………………………………………………..……..46

Importance……………………………………………………………………48

Introduction......................................................................................................49

Page 8: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

vii

CHAPTER Page

Materials And Methods…………………………………………………..…..51

Cells And Viruses………………………………………………………..51

Plasmids And Plasmids Constructions..…………………………………52

Indirect Immunofluorescence Microscopy..……………………………..53

Western Blot Assay.……………………………………………………..54

Glycosidase Treatment.………………………………….........................54

Mice Inoculations………………………………………………………..55

Mv Neutralization Assay...………………………………………………55

DV And YFV Neutralization Assay……………………………………..56

Results………………………………………………………………………..57

Generation And Characterization Of Recombinant Mvs Expressing

Dengue 2 Envelope Antigens…………………………………………….57

Expression Of Dengue 2 E Glycoprotein By Recombinant Viruses…….58

Immunogenicity Of Mv-Dengue 2 Recombinant Viruses……………….59

Generation, Characterization And Immunogenicity Of Mvvac2-

D4(PrMEsol)N Expressing A Soluble Version Of Dengue 4 E

Antigen.......................................................................................................60

Immunogenicity Of Recombinant Mv Vectors Expressing Esol Antigens

From Dengue 2 And 4……………………………………………………62

Cross-Neutralizing Immune Responses In Mv-Susceptible Mice

Inoculated With Mvvac2-D2(PrMEsol)N And Mvvac2-D4(PrMEsol)N..63

Page 9: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

viii

CHAPTER Page

Discussion…………………………………………………………………….65

References…………………………………………………………………….81

4 GENERATION OF A RECOMBINANT MEASLES VIRUS EXPRESSING

FULL-LENGTH DENGUE 4 ENVELOPE GLYCOPROTEINS…....……………...85

Abstract………………………………………………………………………85

Introduction………………………………………………………………….87

Materilas And Methods.…………………………………………………..…89

Cells And Viruses.……………………………………………………….89

Plasmids And Plasmids Constructions…………………………………..90

Indirect Immunofluorescence Microscopy………………………………91

Mice Inoculations………………………………………………………..91

MV Neutralization Assay………………………………………………..92

DV Neutralization Assay………………………………………………..92

Results…………………………………………............................................93

Generation Of Recombinant Divalent MV-DV4 Vaccine Vector………93

Replication Fitness Of Recombinant MV-DV4 Is Equivalent To Parental

MV……………………………………………………………………….94

Recombinant MV-DV4 Induces Anti-Measles And Anti- Dengue 4

Immune Responses…………………………………………………..…..95

Discussion……………………………………………………………………95

References…………………………………………………………………..104

Page 10: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

ix

CHAPTER Page

5 SUMMARY AND FUTURE DIRECTIONS………………………..................106

References…………………………………………………………………....111

COMPREHENSIVE LIST OF REFERENCES…………………………………..........113

Page 11: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

x

LIST OF FIGURES

Figure Page

1.1. Dengue Vaccine Mode Of Action. ……………………………………...................23

1.2. Schematic Representation Of Central Hypothesis...………………………...…......24

2.1. Immunogenicity Of Monotypic Immunity To Dengue 2 And 4……..….………....42

2.2. Cross-Neutralizing Immune Responses In Mice Inoculated With DV1, DV2, DV4,

And YFV ……………………………………………..…….....……………………......43

3.1. Generation And Characterization Of Recombinant Mvs Expressing Dengue 2

Envelope Antigens. ……………………………………………………..……….……..70

3.2. Expression Of Dengue 2 E Glycoprotein By Recombinant Viruses………….……72

3.3. Immunogenicity Of MV-Dengue 2 Recombinant Viruses. ..……………….…...…74

3.4. Generation, Characterization And Immunogenicity Of Mvvac2-D4(Prmesol)N…..76

3.5. Immunogenicity Of Recombinant MV Vectors Expressing Esol Antigens From

Dengue 2 And 4……………………………………………………………….………...78

3.6. Cross-Neutralizing Immune Responses In MV-Susceptible Mice Inoculated With

Mvvac2-D2(Prmesol)N And Mvvac2-D4(Prmesol)N …………………...……………..80

4.1. Generation And Characterization Of Recombinant Mvs Expressing Dengue 4 PrM

And E Antigens……………………………………………………………...…………..99

4.2. Replication Finesses Of Recombinant MV-DV4………………………………….101

4.3. Immunogenicity Of The MV-DV4 Recombinant Virus.……………………….....102

4.4. Sequance Alignment Of TM Regions Of E Protein From DV1-4 And YFV……..103

Page 12: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

1

CHAPTER 1

INTRODUCTION

I-The Measles virus reverse genetics system in the development of multivalent

vaccine options.

Measles virus biology.

Measles virus (MV) is a negative-sense single-stranded RNA virus that belongs to

the Paramyxoviridae family. It is an enveloped, pleomorphic virus with two surface viral

glycoproteins, the Hemagglutinin (H) and Fusion (F) proteins arranged as dimer of

dimers and trimmers, respectively [3]. MV H protein is a type II integral membrane

protein and it is responsible for the viral attachment to cellular receptors. Three proteins

have been indentified as MV cellular receptors, which explain MV tropism. The

complement regulatory protein CD 46 (membrane cofactor protein) was identified as a

cell receptor exclusive for attenuated strains, its role in vivo is unknown. CD 150 or

SLAM (Signaling Lymphocyte Activation Molecule), and Nectin-4 explain viral tropism

for immune competent cells and lower-tract epithelium [4-6]. The fusion protein is a type

I integral membrane protein which is expressed initially in an immature form called F0

and mature after portease cleavage into a disulfide bridged protein with two subunits

called F1 and F2. It is responsible for the triggering of membrane fusion when MV enters

into host cells and when infected cells fuses with neighboring cells to induce synctium

formation [7]. Both MV H and F glycoproteins bind to the matrix (M) protein on the

inner side of the viral membrane through their cytoplasmic tail. M protein thus lines the

inner surface of the viral envelope and plays a role in viral assebly. The MV genomic

Page 13: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

2

RNA is covered by a nucleocapsid (N) protein in tight association with the large RNA-

dependent RNA polymerase (L) and its co-factor, the phosphoprotein (P). MV genome is

infectious only when present in this complex. Full-length MV genomic RNA should be a

multiple of six nucleotides (hexamers) in order to be encapsidated into this RNP

complex[8].

MV replication and transciption. MV N protein is the most abundantly

exppressed protein among measles proteins and plays a critical role in viral replication, as

only encapsidated genomes and antigenomes can replicate. MV L protein is responsible

for the transcription and replication of the genomic RNA and is biologically active in the

presence of the P protein, an essential polymerase cofactor. Replication of MV genomic

RNA is a two-step event. First, the RdRp bind to the leader region at the 3' end of the

genome and synthesize a positive stranded, full-length RNA (antigenomic RNA).

Second, The RdRp uses this anti-genomic RNA as a template to create multiple copies of

the genomic RNA with negative polarity. MV genome transcription is also carried out by

the RdRP and creates transcription gradients of the measles cistrons. MV genes are

organized in independent transcription units with their own cis -active sequences located

at gene boundaries. Short intergenic regions, where the polymerase stops and

reinitializes at intergenic regions, separate each gene. Transcription of cistrons closer to

the genomic 3' end are more abundant; thus the N protein has the higher expression level

while L protein has the lowest. In this way the RdRp modulates the amount of each gene

product. MV replication and transcription is cytoplasmic. MV H and F proteins will be

directed to the endoplasmic reticulum and Golgi to be glycosylated and cleaved to

Page 14: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

3

generate mature forms and then transported to the plasma membrane. The H-F

glycoproteins complex will be assembled on the infected cell surface resulting in fusion

between neighboring cells. Most of the new synthesized virus particles are not released

into the extracellular space, as they transmit to neighboring cells due to fusion. This

fusion of infected cells results in syncytia formation which a sign of measles infection in

cell culture[9].

The rescue of Measles virus from cloned DNA.

MV antigenomic cDNA were obtained from a vial of the Schwarz vaccine using

RT-PCR in order to generate a plasmid called pB(+)MVvac2[10, 11]. The backbone of

this plasmid is PBluescript. Afterwards, a reverse genetic system using either helper virus

or helper cells was developed to rescue (generate) MV from this infectious

complementary antigenomic DNA[12]. The first attempt to rescue MV was attempted

using a helper virus, Vaccinia virus expressing T7 RNA polymerase, T7 driven

transcription at a high levels is necessary to generate a full-length antigenomic copy from

the cloned cDNA. Additionally, infected cells were transfected with the cDNA clonal

copy of the MV genome along with expression plasmids encoding N, P, and L genes also

under the control of the T7 promoter. MV N, P, and L proteins will be used in the

replication of the T7 transcribed genome. However, although the helper virus method is

an effective approach to rescue MVs, it is not the most straightforward strategy to recover

recombinant MV since further intensive purification steps are required. Indeed the

chemical and physical stress used to eliminate vaccinia infectivity can inactivate MV. A

modified method was introduced by Radecke to rescue MV using Helper cells (293-3-

Page 15: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

4

46). Those engineered cells expresses P, N proteins, and T7 polymerase in a constitutive

manner. In this approach, Helper cells are transfected with the RDRP expressing plasmid

pEMC-La and the full-length cDNA infectious clone. Thus, expressesion of the L

protein and MV antigenomes flanked by T7 promoter and a precise terminator will start

MV replication and transcription. In helper cells cytoplasm T7 polymerase will transcript

MV anti-genomic RNA that will form the RNP complex with N, P, and L protein. The

RdRp will then generate the MV genomic RNA that will be used the express all of MV

proteins. The progeny of rescued MV is propagated by overlaying the transfected rescue

cells with a cell line that support MV replication such as Vero cells expressing the

receptor SLAM[13].

Genomic modification of MV- insertion of foreign coding sequences-

To facilitate the cloning of foreign coding sequence into MV vectors, additional

transcription units (ATU) need to be inserted into the intergenic regions of the MV

antigenomic sequence. Inserting a multicloning cassette of unique restriction sites is

convenient to facilitate further cloning. The insertion of the ATUs in intragenic regions

allows the direct the transcription by RdRp of the foreign coding sequence. ATUs were

first inserted downstream of the P, H or L genes, in order toallow the possibiity of

modulating expression levels. Afterwards, a second generation of MV vectors directing

the expression of genes inserted downstream the N gene (in position 1710) was generated

to drive protein expression at almost maximum levels. The insertion of open reading

frames coding fluorescent markers have been used to test the functionality of the inserted

ATUs and also track the viral infection in vivo [13].

Page 16: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

5

Multivalent recombinant measles vaccines.

The attenuated MV vaccine is one of the most stable, safe, and effective human

vaccines. Vaccination using measles vaccine induces life-long protection against MV

infection. This strong and long-term protection could be extended if recombinant MV

expressing of other pathogens’ antigens are obtained and amplified. The MV vaccine has

been used for more than five decades, with a well-established production and distribution

network, therefore the cost of production of recombinant multivalent MV vaccine would

be relatively cheaper than other vaccines candidates. The MV vaccine platform is a safe

choice to deliver foreign antigens, as it replicates exclusively in the cytoplasm of the

infeted cells, thus the danger of integration into human genome is excluded. Also, MV

vaccine vectors have the capacity to carry and express foreign inserts without restriction

due to its helical capsid structure.

The capacity of MV genome to carry and expresses large inserts and the stability

of the expression make it a target of intense research to generate a variety of recombinant

multivalent vaccines[14-18]. The first multivalent MV-based vaccine was generated to

induce protective MV and hepatitis B virus (HBV) immune responses by expressing the

small surface antigen (HBsAg)[19]. The coding sequence was inserted downstream P, H,

and L genes. In HuCD46Ge-IFNarKO mice, the three recombinants MV induced

antibodies responses against both MV and HBV in which the vectors with the insertion

downstream P elicited the highest and the vectors with the insertion downstream L

elicited the lowest humoral immune responses against HBV. At that time, the P cassette

vector was the strongest expressing and generated an optimal immune response against

Page 17: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

6

HBV. Rhesus monkeys immunized with the high-expression vector, with the HBsAg

insertion downstream of the P gene, had the most robust immune response against HBV

and remain protexted against a mealses virus challenge, providing proof -of-principle for

the divalent vaccine strategy, where a pediatric measles virus vaccine vector.

Furthermore, AIDS is arguably one of the most important infectious disease worldwide

against which a vaccination option has not been approved yet. HIV and MV efficiently

infect monocytes, dendritic cells, and macrophages and induce immune suppression.

Thus, an MV-derived vector has the ability to deliver HIV protein antigens to the same

target cells than HIV does, resulting in strong humoral and cellular immune responses

against HIV and MV. To test the immunogenicity of MV encoding both an HIV Gag

antigen inserted downstream the MV P gene, as well as an Env (gp 140) antigen

downstream H gene, macaques were immunized twice, two and a half months apart[20].

All vaccinated animals developed immune responses against measles and HIV. This

finding makes the MV-based HIV vaccine a promising candidate to be tested in human to

determine its safety and immunogenicity[21]. Chikungunya virus is a mosquito-

transmitted alphavirus causing millions of cases of severe polyarthralgia with no vaccine

currently available. Recently, a recombinant live attenuated measles vaccine (MV)

expressing Chikungunya virus (CHIKV) virus-like particles by inserting capsid and

envelope structural proteins coding sequences downstream the P position[22] was tested

in HuCD46Ge-IFNarKO mice with two doses of MV-CHIKV, ranging from 10^3 to

10^5 TCID50, given one month apart. High neutralizing antibodies titers were induced

(PRNT50= 450–4050 and PRNT90= 50–450). The MV- based CHIKV has been the first

Page 18: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

7

recombinant MV multivalent vaccine to progress to phase I trial based on

immunogenicity studies in mice. In this phase I clinical trial, healthy men and women

aged 18–45 years received either one of three doses of the measles-virus-based candidate

vaccine 1•5 × 10³, 7•5 × 10³ or 3•0 × 10⁰ TCID50 and then received a booster injection

on either day 28 or day 90 after the first vaccination. The primary endpoint was the

presence of neutralizing anti-chikungunya antibodies on day 28, as assessed by 50%

plaque reduction neutralization test. The candidate vaccine raised neutralizing antibodies

in all dose cohorts after the second vaccination dose resulting in a 100% seroconversion

for all participants. The immunogenicity of the candidate vaccine was not affected by

pre-existing anti-measles immunity which make it a promising measles-virus-based

candidate vaccine for use in human [23].

II- The quest for an optimal, Pediatric Dengue virus vaccine.

Epidemiology of dengue virus (DV).

In the last 60 years, the incidence of clinical cases of dengue infections reported

to WHO has increased 30-fold, with a much-increased geographic range, including the

expansion from predominantly urban to rural settings with a tremendous medical

importance in tropical and sub-tropical areas[24]. Dengue infections are endemic in large

parts of Africa, Asia, and Latin America with approximately 3.5 billion people living in

endemic countries. With over 500,000 estimated hospitalizations annually due to dengue

infections, of which 12000 cases are fatal, the tremendous burden of health care

represented by dengue virus has a high impact on health costs[25]. Most frequently,

Page 19: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

8

Dengue infection results in a spectrum of clinical manifestation ranging from

asymptomatic to dengue fever (DF), which is characterized by flu-like-symptoms, the

patient will fully recover with no hospitalizations. Severe dengue infections might cause

life-threatening dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS)[26].

In DHF, patients experience hemorrhagic tendencies evidenced by spontaneous bleeding

from the mucosa, gastrointestinal tract, and other locations, as weel as thrombocytopenia

and evidence of plasma leakage due to increased vascular permeability. As proof for

capillary hemorrhage, patients with a high tendency to hemorrhage present a positive

tourniquet sign. DSS is characterized by a rapid or a weak pulse and narrow pulse

pressure. Hemodynamically, the shock syndrome is associated mainly with a generalized

vasodilation that causes a reduction in peripheral resistance. This severe dengue infection

is life threatening primarily in patients younger than 15 years old[27, 28].

Dengue virus is the most important arbovirus disease that is transmitted to human

by mosquito bites. Two mosquito vectors transmit dengue virus, Aedes.aegypti is the

predominant vector and Aedes.albopictus can sustain transmission in some endemic

areas[29]. Mosquitos are infected after the ingestion of infectious blood meal. DV infects

and multiplies in midgut epithelial cells. The viral infection gets amplified by infected

other mosquitoes’ cells, when it reaches the epithelial cells of the salivary gland the

mosquitos will be capable of transmitting the virus to human when feeding. The period

comprehended between the transmission of dengue virus from an infected human to the

mosquito and the subsequent amplification of dengue virus in mosquito’s cells is called

the extrinsic incubation period. Once the infected mosquitoes transmit the virus to a

Page 20: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

9

human host, the virus will replicate into resident cells at the site of the mosquito bite. The

period between human infection and the onset of symptoms is known as intrinsic

incubation period[30].

The pathogenesis of Dengue infection.

As mentioned, DV infects human through the cutaneous route. After the mosquito

bites, the virus crosses skin epithelial cells and reaches the subepithelial tissues[30, 31].

Epidermal dendritic cells, known as Langerhans cells, are the primary targets of

infection. DV will be amplified in dendritic cells. Upon infection, dendritic cells get

activated and migrate to neighbor lymph nodes where monocytes and macrophages are

recruited and they get infected in a process called lymphatic spread of DV[32]. The

infected macrophages and monocytes will circulate through the blood stream, which

constitutes the primary viremia. Once the virus gets into the blood stream the viral

infection will be spread to other organs. Intense DV replication in targeted host cells

results in secondary viremia with an intensity of 10^5 to 10^6 dengue-infectious units per

ml at its peak. This period includes infection of blood-derived monocytes, myeloid

dendritic cells and tissular macrophages[33]. Infected macrophages will be major hosts in

the liver, spleen, and bone marrow stromal cells, while kidney, lung, and the central

nervous system infection by dengue is one of the main events associated with DHF and

DSS, where the viremia is 10 to 100 fold greater than in DF[26]. In DHF, infected

macrophages infect both hepatocytes and Kupffer cells resulting in necrosis and

apoptosis in liver. As a result, decreases in the liver function include the release of toxic

products, reducing the synthesis of coagulation factors, declining the level of platelets

Page 21: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

10

and the development of coagulopathy disorder[34]. The replication of DV in bone

marrow stromal cells causes suppression of haemopoiesis that may cause lymphopenia

and leukopenia. In conjunction, severe thrombocytopenia and platelet dysfunction,

associated with eventual loss of vascular stability, can occur. Thus, increasing capillary

fragility and gastrointestinal mucosal bleeding is a characteristic of DHF. Furthermore, a

severe infection of endothelial cells causes plasma leakage as a feature of DSS[33].

Dengue virus biology.

Dengue is an enveloped, positive sense, single-stranded RNA virus of the

Flaviviridae family with an icosahedral scaffold of 90 glycoproteins envelope (E) dimers.

The DV complex has four serotypes (DV 1-4), phylogenetically the four DV serotypes

are diatintive and always cluster conspicuosly within the mosquito-borne flaviviruses

branch. The amino acid sequence of the envelope protein from the four serotypes are

homologous by 63-68%, DV 3 and 1 sequence is more related to each other than to the

other two serotypes[35]. The positive polarity genomic RNA is about 10.6 kbp with a

type I 5’ cap and a 3’ end lacking a polyadenylate tail. The genome encodes a single

long open reading frame (ORF) flanked by 5’ and 3’ noncoding regions including

structural and non-structural proteins which includes, capsid (C), precursor membrane

(PrM), envelope (E), nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and

NS5)[31]. The capsid (C) protein is a highly basic alpha-helical protein. The nascent C

contains a hydrophobic anchor at their C-terminal that serves as a signal peptide for

endoplasmic reticulum (ER) translocation of PrM and the first signal sequence to be

translated by cellular ribosomes. This hydrophobic domain is cleaved by viral proteases

Page 22: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

11

to release mature C proteins, which fold into compact dimers with each monomer

containing alpha helices. These will then bind to the genomic RNA and interact with the

membrane surface to form the DV nucleocapsid or the viral core. The glycoprotein

precursor of M protein contains glycosylation sites at the N –terminal region while the C-

terminal has the transmembrane domain (TM) that act as ER- retention signals to keep

the protein associated with ER membrane. The PrM protein is about 26 KDa present on

the surface of the immature virus and responsible of forming a cap to cover the fusion

loop of the E protein to prevent this from fusion during transit through the secretory

pathway. During DV maturation, the PrM will be cleaved by cellular proteases to release

the glycosylated, N-terminal Pr fragment from the small, membrane anchored M protein

which is about 9 KDa and function as a scaffold to maintain E protein dimers folded

correctly[36, 37]. The envelope protein is type I integral glycoprotein and the major

protein on the surface of Dengue virions, This glycoprotein of about 53 KDa mediates

host cell receptor binding and membrane fusion[38]. Each E protein monomer contains

two helix transmembrane regions, which are about 50 a.a called stem and anchor regions

and contain an ER retention signal, which is responsible for association of the E protein

with ER membrane. The ectodomain is structurally divided into three domains that are

joined by a flexible junction. Domain I is the middle domain and participate in the

conformational changes induced during viral fusion. Domain II contains a hydrophobic

fusion loop that mediates the viral- cell membrane fusion. Domain III has an

immunoglobulin-like folding pattern and is responsible for attachment and receptor

binding onto host cells[39, 40]. Non-structural protein 1 (NS1) is present on the surface

Page 23: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

12

of infected cells in form of highly stable homodimers or is secreted into the extracellular

space. The function of this protein in viral replication is not known. NS2A can act as an

interferon antagonist and assist DV evasion of the interferon inhibition of viral

replication. One of the most important nonstructural proteins is NS3 which carries out

multiple functions including serine-like protese, RNA helicase- NTPase and RTPase

enzymatic activities. The protease activity of NS3 depends on the NS2B that is a cofactor

forming an NS2B-NS3 serine protease complex. NS4A and B are small hydrophobic

proteins which acts as antagonist to block type I interferon-signaling pathways that are

essential to enhance DV replication. The largest (104 KDa) and most conserved protein

among DV serotypes is the NS5 protein that is the RNA-dependent RNA polymerase

(RdRp)[36, 41-44].

The basic steps of DV replication are attachment to cell surface; internalization;

release of the viral nucleocapsid and transfer of viral RNA into the cytoplasm; translation

of the viral polyprotein; replication of genomic RNA; assembly, maturation of virions,

and ultimately the release of mature viruses from the cells. Receptor recognition and

attachment of E glycoprotein and concentration of dengue virions on the surface of host

cells initiate viral infection by receptor-mediated endocytosis of the mature DV. The

plasma membrane vesicles bud into the cytoplasm and deliver the virus particle to early

endosomes in which the viral membrane- endosome membrane fusion occurs as the pH

dropps. The fusion is mediated by the hydrophobic sequence present in the fusion peptide

resulting in the generation of a pore in the endosome membrane that allows the release of

nucleocapsid into the cytoplasm. The RNA is then translated into a polyprotein and

Page 24: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

13

directed into the endoplasmic reticulum (ER). Signal sequences within polyprotein

translocate the E, PrM, and NS1 proteins into lumen of the ER, while the C protein, NS3

and NS5 are released into the cytoplasm. The RdRp is used to replicate the genomic RNA

in the cytoplasm which then migrate to the ER where the assembly of the virus take place

once there are enough glycoprotein copies. The assembled immature virus particles

transit from ER through trans-Golgi network to the cell surface. During the maturation,

PrM protein is cleaved by cellular furin protease to Pr and M and will remain associated

with the virions, once the virions are released into the extracellular space the Pr peptide

will be released and virions will be on its mature infectious form where the E protein is

arranged in a 90 homodimer lattice liying upon the virus membrane and completely

covering the small M protein. DV replication can generate subviral particles that contain

only PrM and E, these particles are less dense than the virions and have significant

importance in vaccine development[31, 38, 40, 45, 46].

Immune response and immunopathogenesis of Dengue virus infection.

After the initial bite of an infected mosquito, host cells are the first line to mount

innate immune response to DV infection by ultimately activating type I interferon.

Infected cells posses cytoplasmic sensors for the replication of DV genomic RNA such as

retinoic acid inducible gene I (RIG-I), which triggers the anti-viral interferon responses in

infected cells, transfering signals to neighborhood cells to clear the infection[47]. DV has

evolved to escape the first line of innate defense through blocking the interferon system

signaling pathways using NS2A, NS4A, NS4B, and NS45[48, 49]. The cytokine release

set up a state of tissue inflammation that is associated with dilation of local blood vessels

Page 25: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

14

and permeability increase in endothelial cells, allowing the recruitment of other immune

cells such as neutrophils and monocytes out of the blood vessel and into infected tissue

and active phagocytosis of DV present in the extracellular space. The most important

event of the innate immune response is the recruitment of natural killer (NK) cells that

are responsible for recognizing and killing infected cells. At this point, DV infection may

progress and spread into innate immune cells which will migrate to local lymph nodes for

subsequent presentation of DV antigen to naïve T and B-lymphocytes. Activated

lymphocytes coordinate the adaptive immune response which has two goals; the blocking

of viral entry by antibodies secreted by plasma cells (from mature B cells); and the

elimination of infected cells by cytotoxic T cells (CTL). CTL recognize DV antigenic

epitopes in complex with MHC class I molecules on the surface of infected cells where

CTL can be activated to secrete cytotoxic protein products including granzymes and

perforin to induce apoptosis of infected cells [50-53]. The activity of CTL is part of the

cellular immune response to clear DV in infected patients, however the most effective

immune response to curtail dengue infection is the humoral response mediated by

secreted antibodies. Effector functions of anti-DV antibodies includes neutralization of

DV; opsonization of DV coated by antibodies; complement activation; and antibody

dependent cellular cytotoxicity[54, 55]. The primary role of the antibodies is to block the

viral entry into target cells by recognizing and binding DV glycoproteins on the surface

of virions. Anti-E antibodies are responsible for neutralization and inhibition of DV

infection by binding to E glycoprotein and blocking DV attachment and entry. It has

been shown that the recognition and binding of neutralizing monoclonal antibodies to

Page 26: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

15

exposed regions in the viral envelope causes the destabilization of the infectious particles

and interfere with viral entry[56]. Thus, the elicitation of neutralizing antibodies through

vaccination has been the most desired strategy to curtail the spread of mosquito-born

flavivirus[57]. For instance, the attenuated Yellow Fever virus (YFV) strain 17D has

been in use for more than 60 years. A single dose elicits seroconversion in 100% of

vaccinees. Therefore, neutralizing immunity is the most clear and documented correlate

of protection against dengue virus. Protective immunity against secondary infection of

DV is one of the most significant consequences of adaptive immunity by generating a

population of memory B-lymphocytes that produce rapid and vigorous immune responses

in secondary infection due to the same serotype. Individuals who have been infected with

one dengue serotype have long-term protection against that specific serotype and short-

term protection against the other three serotypes of DV.

The immune response to secondary heterotypic dengue infection can result in

unfavorable clinical outcomes such as DHF and DSS, which is known as

immunopathogenesis of DV infection. There is some evidence suggesting that specific

antibodies play a role in viral pathogenicity, such as the increased risk to develop DHF

and DSS during primary infection in infants whom are born to mothers with established

herteotypic immunity to DV. The tropism of DV for monocytes and dendritic cells, both

expressing Fc receptor for the heavy of IgG creates the opportunity for DV specific

antibodies to enhance the viral entry in a phenomenon called antibody- dependent

enhancement (ADE). In ADE, cells covered by Fc receptor will up take the virion-

antibody complex more efficiently than Dengue viral entry through specific cell-

Page 27: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

16

receptors. This results in increasing of the viral loads and enhancement of the disease.

Therefore, development of dengue vaccine requires the generation of tetravalent vaccine

formulations to avoid the risk of ADE development in vaccinees[34, 55, 58-61].

Dengue vaccine candidates.

The first attempt to generate a Dengue vaccine dates from 1945 by A.Sabin who

tried to produce an attenuated DV bypassing the virus in mouse brain; the virus was

reactogenic and caused a rash in human volunteers[62]. Since this, there have been

efforts to develop a dengue vaccine; the reason why dengue is one of the most difficult

pathogens to develop a vaccine against is the difficulty to induce a balanced protection

without viral interference following the simultaneous vaccination against the four

serotypes[54]. Several vaccines candidates are under different stages of development

including six advanced candidates that are currently in various stages of clinical

development with vaccine trials in phases I–III[63]. Three candidates are in phase I, a

recombinant subunit vaccine based on the DV premembrane and truncated envelope

proteins (DV-PrM- 80% E) via antigenic expression in Drosophilla S2 cell system

(V180) by Merck[64]. A monovalent plasmid DNA expressing DV 1 PrM and E proteins

under control of the human cytomegalovirus promoter (D1ME 100) by Naval Medical

Research Center (NMRC)[65, 66]. And a tetravalent purified inactivated vaccine (TDEN

PIV) evaluated jointly by GSK and Walter Reed Army Institute of Research (WRAIR)

and tested in flavivirus naïve adults (USA) and dengue primed population (Puerto Rican)

with no adjuvants or with different type of adjuvants respectively[67, 68]. Two

Page 28: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

17

candidates have advanced to the phase II trials, TV003/TV005, and DENVax that show

promising results to progress to phase III.

The National Institute of Allergy and Infectious Disease (NIAID) developed a

tetravalent vaccine based on wild-type strains with targeted mutation in the 3’

untranslated region to attenuate the virus. Vaccine virus serotypes 1, 3, and 4 are based

on whole attenuated virus, while serotype 2 is a recombinant virus based on attenuated

serotype 4 psuedotyped with serotype 2 PrM and E glycoproteins. TV003 and TV005 are

identical vaccine candidates except for the dosage of DV2 component, where each

component was given at a dose of 3 log10 PFU as a single subcutaneous dose with the

exception of the DENV2 component of TV005, which was given at a dose of 4 log10

PFU. To determine the safety and the immunogenicity of TV003 and TV005 in 168

flavivirus-naive adults, a single dose of TV003 induced seroconversion rates to DENV1–

4 of 92%, 76%, 97%, and 100%, respectively. Seventy-four percent of TV003 recipients

mounted a tetravalent response. A single dose of TV005 induced seroconversion rates to

DENV1–4 of 92%, 97%, 97%, and 97% , respectively and an overall tetravalent response

in 90% of recipients. A phase II study in Thailand started in early 2015 to examine safety

and immune responses of two doses of TV003 at 0 and 6 months in adults and children.

In a phase II, TV005 is currently being evaluated in an age de-escalation trial in Thailand.

A phase II study of TV003 in Brazil started in October 2013 in collaboration with the

Butantan Institute. Safety and immunogenicity of one dose of TV003 in dengue naïve and

dengue primed individuals will be evaluated with results completed by December

2018[69-73].

Page 29: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

18

Takeda is also developing a live recombinant tetravalent vaccine DENvax (TDV)

including a mixture of a live attenuated DV2 and chimeric DV1, 3, 4 based on the

attenuated DV2 backbone[74]. The wild type DV2 was attenuated by passaging in

primary dog kidney (PDK) cells, the DEN-2-PDK-53 vaccine, which is further attenuated

by a mutation in NS3 gene and used as backbone for the rest of the vaccine component

by replacing the PrM and E of DV2 with ortholog genes from the respective virus wild

type serotypes. There have been several ongoing and completed phases II and I trails in

both endemic and dengue naïve populations. An ongoing phase II study of 344 children

and adults assessing the safety and immunogenicity of the vaccine in Colombia, Puerto

Rico, Singapore and Thailand started in 2011. In late 2014 a phase II study in 1800

children in Asia and Latin America commenced to examine immunogenicity of a 3 dose

schedules[75-77].

CYD-TDV (Dengvaxia®) is a tetravalent, live attenuated viral vaccine developed

by Sanofi Pasteur and licensed in five endemic countries: México, Philippines, Brazil, El

Salvador, and Paraguay. CYC-TDV is composed of 4 live attenuated chimeric viral

recombinants representing dengue 1, 2, 3, and 4 each one obtained by replacing the genes

encoding prM and E proteins in the attenuated YFV 17D genome with the corresponding

genes from the 4 serotypes of wild type dengue[78]. It is approved to prevent severe

dengue disease caused by DV in individuals 9 through 45 or 60 years of age, with a clear

contraindication to be used in children aged 2-5 years old due to safety concerns, as

evidenced after phase 3 clinical trials. The efficacy of the CYD14 included all children of

all ages where vaccine efficacy (VE) in young children was 33.7% compared to VE in

Page 30: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

19

adult 74.3%[79, 80]. Vaccine efficacy measures the proportionate reduction in cases

among vaccinated person by calculating the risk of disease among vaccinated and

unvaccinated persons and determining the percentage reduction in risk of disease among

vaccinated persons relative to unvaccinated persons. Data from endemic and non-

endemic countries show an increase risk of hospitalization and severe dengue at later

follow up periods as a result of vaccination at age younger than 6 years (58

hospitalization cases in vaccinees in comparison with 23 among control recipients). Thus,

it is documented the possibility that vaccination may be not sufficient or even increase

the risk of hospitalization and severe dengue outcome in young children who are

seronegative at the time of first vaccination if this occurs at 2-6 years of age[81, 82].

Several hypotheses have been suggested to explain this result in seronegative young

children[83], including that the vaccine act as a silent natural infection that prime

seronegative vaccinees to experience a secondary-like infection upon their first exposure

to dengue virus (Figure 1.1)[2]. Following vaccination of individuals with no pre-existing

immunity, primary infection acts like secondary infection while in persons who were

vaccinated after experiencing one natural infection behave like a natural tertiary

infection. Under these conditions, very young individuals who were vaccinated while

seronegative are at higher risk for dengue disease upon infection.

Page 31: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

20

Central Hypothesis

The recent phase III and II of the CYD-TDV, DENvax, and TV003/5 show

efficacy in terms of protection against severe dengue disease in old children and adult.

However, it has been documented the possibility that vaccination may be not sufficient or

even increase the risk of hospitalization and severe dengue in young children who are

seronegative at the time of first vaccination when this occurs at 2-6 years of age. The

need to protect young infants from dengue is imperative. The disease is more severe at

this age, and the intense virus replication can be the reservoir to sustain an epidemic

outbreak. Therefore, closing the gap between the timing of the current vaccine and risk of

severing Dengue infection is critical. The enhanced efficacy of the CYD-TDV vaccine in

a seropositive recipient with average vaccine efficacy of 74.6% in seropositive and only

34.5% in seronegative, proves that DV immune responses and protection efficacy is

majorly boosted when the recipient’s immune system is primed against flaviviruses due

to prior exposure to DV or other flavivirus. In this dissertation, we explored an

alternative pediatric vaccine strategy based on multivalent Dengue-Measles

immunization, to prime DV or other representative flaviviral neutralizing immune

responses, reducing the window of DV infection risk. We specifically hypothesized that

recombinant measles virus expressing Dengue 2 and 4 antigens would successfully

induce neutralizing responses against DV2 and 4 and prime cross-neutralizing responses

against other closely related or distance flaviviruses (Figure1.2). The goal of this

dissertation is to develop the concept for a measles vaccine viral vector (MVvac2) to

express the pre-membrane (prM) and the most immunogenic form of the major envelope

Page 32: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

21

(E) from DV2 and DV4. Also to test homologous neutralizing responses and cross-

neutralizing immune responses elicited by single-component or combined recombinant

MV vaccines in MV-susceptible mice. I expect that this research would lay the

foundation for future experimentation in more relevant primate models.

Chapter Organization

Chapter 2 of this Dissertation is entitled “Broad heterologous protection in

flavivirus infections." describes homologous neutralizing immune responses to DV2 or

DV4 and heterologous neutralizing responses to DV1 and YFV 17 D (attenuated).

Additionally, in this chapter we show the correlation between titrating neutralizing

antibodies by 50% plaque reduction neutralization titer (PRNT50) and the logarithmic

neutralization index (LNI).

Chapter 3 of the dissertation is entitled "Generation of a complementary anti-

dengue immunization strategy based on a recombinant measles virus vaccine cocktail." It

describes the generation of three recombinant measles. Two of which expresses PrM and

two forms of DV2 E protein: full length or truncated (90% of E) and one recombinant

measles expressing PrM and truncated E of DV4. The two recombinant measles

expressing DV2 antigens replicate with equal efficiency to their parental strain. The

expression of DV2 antigens is evident by indirect immunofluorescence and western blot.

While both vectors are immunogenic, the virus expressing truncated E showed a

distinctive more immunogenic profile in MV-susceptible small animal model. Based on

that, we generated the recombinant measles expressing truncated E protein of DV4 and

Page 33: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

22

showed that this vector is immunogenic as well. Finally, we combined the recombinant

measles expressing truncated E protein of DV2 and DV4. The cocktail of recombinant

measles generates homologous and heterologous immune responses against DV2 and 4,

and DV1 and YFV, respectively.

Chapter 4 of the dissertation is entitled "Generation of a recombinant measles

virus expressing full-length Dengue 4 envelope glycoproteins”. It describes the

generation of recombinant measles expressing full-length DV4 E protein. This virus

replicates with equal efficiency the parental strain in two cell lines. The expression of

DV4 antigens is evident by indirect immunofluorescence. Finally, the immunogenicity of

the virus is tested in MV susceptible mice. We documented that, despite clear differences

in DV4 E transmembrane region, immunogenicity against vectored DV4 Esol antigen

followed the same principle as the MV-vectored DV2 Esol.

Chapter 5 of the dissertation is entitled "Summary and future directions." It

discusses the implications of my findings presented in this dissertation as well as avenues

for future research.

Page 34: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

23

Figure 1.1. Hypothesized mode of action of the current Dengue vaccine. Red arrow indicates exposure to dengue infection. The height of the boxes illustrates the chance of severe dengue of occurring. The syringe symbol shows the timing of vaccination. Upper panel shows the outcome of multiple dengue infections in naïve, unvaccinated individuals over their lifetime. Middle panel shows the impact of the vaccine on increasing the risk of severe disease in seronegative vaccinees. In this model, vaccine acts as natural silent primary infection, which put the vaccinated individuals at high risk to develop severe dengue upon exposure to wild type dengue virus. Lower panel shows vaccine-induced protection in seropositive individuals. Vaccination confers long-lived high-level protection against disease in individual who are vaccinated after a single flavivirus infection[2]

Page 35: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

24

Figure 1.2. Schematic representation of our central hypothesis. A. Cartoon representation of a divalent measles-dengue vaccine concept and the expected neutralizing antibody responses. B. Proposed mode of action for a MV-Dengue vaccine. The syringe symbol shows the timing of vaccination. Seronegative infants receive a cocktail of MV-DV vaccine at one year old to prime their immune systems. At 18 month of age they receive the CYD-TDV, by this time all the vaccinees will be seropositive, closing the gap of opportunity for a wild type infection to cause severe Dengue.

Dengue2'Glycoproteins''

Measles'glycoproteins'

An56Measles'an5body''

An56Dengue'an5body'

Dengue4'Glycoproteins''

A'

B'!"#$%&'%()*%+",$%&-*."*/&

01231&$"44-*%&4)45/"-6&

'%()7)8,$-/9&

:%/("$"6%*/&31&1"44-*%&

;<&=)*/>&&)6?&

;@&=)*/>&&)6?&

8-A&=)*/>8&

B(-="(9&-*.%4,)*&!)&8%$%(%&3%*+C%& D)*+&6-.%&7()/%4,)*&

Page 36: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

25

REFERENCES

1. Stefan Flasche1*, M.J., Isabel Rodríguez-Barraquer 2*, Laurent Coudeville3*,

Mario Recker4*,, et al., Comparative modelling of dengue vaccine public health impact, 2016, World Health Organization.

2. Griffin, D.E. and M.B. Oldstone, Measles: history and basic biology. Vol. 329.

2008: Springer Science & Business Media. 3. Erlenhoefer, C., et al., CD150 (SLAM) is a receptor for measles virus but is not

involved in viral contact-mediated proliferation inhibition. Journal of virology, 2001. 75(10): p. 4499-505.

4. Naniche, D., et al., Human membrane cofactor protein (CD46) acts as a cellular

receptor for measles virus. Journal of virology, 1993. 67(10): p. 6025-32. 5. Muhlebach, M.D., et al., Adherens junction protein nectin-4 is the epithelial

receptor for measles virus. Nature, 2011. 480(7378): p. 530-3. 6. Watanabe, S., et al., Mutant fusion proteins with enhanced fusion activity promote

measles virus spread in human neuronal cells and brains of suckling hamsters. Journal of virology, 2013. 87(5): p. 2648-59.

7. Delpeut, S., et al., Host factors and measles virus replication. Current opinion in

virology, 2012. 2(6): p. 773-83. 8. Abe, Y., et al., Characteristics of viruses derived from nude mice with persistent

measles virus infection. Journal of virology, 2013. 87(8): p. 4170-5. 9. Reyes-del Valle, J., et al., Broadly neutralizing immune responses against

hepatitis C virus induced by vectored measles viruses and a recombinant envelope protein booster. Journal of virology, 2012. 86(21): p. 11558-66.

10. Radecke, F. and M.A. Billeter, Appendix: measles virus antigenome and protein

consensus sequences. Current topics in microbiology and immunology, 1995. 191: p. 181-92.

11. Radecke, F., et al., Rescue of measles viruses from cloned DNA. The EMBO

journal, 1995. 14(23): p. 5773-84. 12. Sidhu, M.S., et al., Rescue of synthetic measles virus minireplicons: measles

genomic termini direct efficient expression and propagation of a reporter gene. Virology, 1995. 208(2): p. 800-7.

Page 37: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

26

13. Brandler, S., et al., Pediatric measles vaccine expressing a dengue antigen induces

durable serotype-specific neutralizing antibodies to dengue virus. PLoS neglected tropical diseases, 2007. 1(3): p. e96.

14. Brandler, S., et al., Pediatric measles vaccine expressing a dengue tetravalent

antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine, 2010. 28(41): p. 6730-9.

15. Brandler, S. and F. Tangy, Recombinant vector derived from live attenuated

measles virus: potential for flavivirus vaccines. Comparative immunology, microbiology and infectious diseases, 2008. 31(2-3): p. 271-91.

16. Despres, P., et al., Live measles vaccine expressing the secreted form of the West

Nile virus envelope glycoprotein protects against West Nile virus encephalitis. The Journal of infectious diseases, 2005. 191(2): p. 207-14.

17. Lorin, C., et al., A paediatric vaccination vector based on live attenuated measles vaccine. Therapie, 2005. 60(3): p. 227-33.

18. del Valle, J.R., et al., A vectored measles virus induces hepatitis B surface antigen

antibodies while protecting macaques against measles virus challenge. Journal of virology, 2007. 81(19): p. 10597-605.

19. Guerbois, M., et al., Live attenuated measles vaccine expressing HIV-1 Gag virus

like particles covered with gp160ΔV1V2 is strongly immunogenic. Virology, 2009. 388(1): p. 191-203.

20. Lorin, C., et al., A single injection of recombinant measles virus vaccines

expressing human immunodeficiency virus (HIV) type 1 clade B envelope glycoproteins induces neutralizing antibodies and cellular immune responses to HIV. Journal of virology, 2004. 78(1): p. 146-157.

21. Brandler, S., et al., A recombinant measles vaccine expressing chikungunya virus-

like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine, 2013. 31(36): p. 3718-3725.

22. Ramsauer, K., et al., Immunogenicity, safety, and tolerability of a recombinant

measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. The Lancet infectious diseases, 2015. 15(5): p. 519-527.

23. Guzman, M.G., et al., Dengue: a continuing global threat. Nature Reviews

Microbiology, 2010. 8: p. S7-S16.

Page 38: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

27

24. Gubler, D.J., Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends in microbiology, 2002. 10(2): p. 100-3.

25. Gubler, D.J., Dengue and dengue hemorrhagic fever. Clinical microbiology

reviews, 1998. 11(3): p. 480-96. 26. Halstead, S.B., Observations related to pathogensis of dengue hemorrhagic fever.

VI. Hypotheses and discussion. The Yale journal of biology and medicine, 1970. 42(5): p. 350-62.

27. Nimmannitya, S., Clinical spectrum and management of dengue haemorrhagic

fever. The Southeast Asian journal of tropical medicine and public health, 1987. 18(3): p. 392-7.

28. Hardy, J., Susceptibility of vector Mosquitoes, in The arboviruses: Epidemiology

and ecology, T. Monath, Editor 1988, CRC Press: Boca Raton, FLA. p. 87-126. 29. Wu, S.J., et al., Human skin Langerhans cells are targets of dengue virus

infection. Nature medicine, 2000. 6(7): p. 816-20. 30. Bartenschlager, R. and S. Miller, Molecular aspects of Dengue virus replication.

Future microbiology, 2008. 3(2): p. 155-65. 31. Ho, L.J., et al., Infection of human dendritic cells by dengue virus causes cell

maturation and cytokine production. Journal of immunology, 2001. 166(3): p. 1499-506.

32. Vaughn, D.W., et al., Dengue viremia titer, antibody response pattern, and virus

serotype correlate with disease severity. The Journal of infectious diseases, 2000. 181(1): p. 2-9.

33. Blackley, S., et al., Primary human splenic macrophages, but not T or B cells, are

the principal target cells for dengue virus infection in vitro. Journal of virology, 2007. 81(24): p. 13325-34.

34. Henchal, E.A. and J.R. Putnak, The dengue viruses. Clinical microbiology

reviews, 1990. 3(4): p. 376-96. 35. Lobigs, M., Flavivirus premembrane protein cleavage and spike heterodimer

secretion require the function of the viral proteinase NS3. Proceedings of the National Academy of Sciences of the United States of America, 1993. 90(13): p. 6218-22.

Page 39: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

28

36. Zhang, Y., et al., Structures of immature flavivirus particles. The EMBO journal, 2003. 22(11): p. 2604-13.

37. Stiasny, K., et al., Molecular mechanisms of flavivirus membrane fusion. Amino

acids, 2011. 41(5): p. 1159-63. 38. Huerta, V., et al., Characterization of the interaction of domain III of the envelope

protein of dengue virus with putative receptors from CHO cells. Virus research, 2008. 137(2): p. 225-34.

39. Stiasny, K. and F.X. Heinz, Flavivirus membrane fusion. The Journal of general

virology, 2006. 87(Pt 10): p. 2755-66. 40. Egloff, M.P., et al., An RNA cap (nucleoside-2'-O-)-methyltransferase in the

flavivirus RNA polymerase NS5: crystal structure and functional characterization. The EMBO journal, 2002. 21(11): p. 2757-68.

41. Munoz-Jordan, J.L., et al., Inhibition of interferon signaling by dengue virus.

Proceedings of the National Academy of Sciences of the United States of America, 2003. 100(24): p. 14333-8.

42. Perera, R. and R.J. Kuhn, Structural proteomics of dengue virus. Current opinion in microbiology, 2008. 11(4): p. 369-77.

43. Zhou, Y., et al., Structure and function of flavivirus NS5 methyltransferase.

Journal of virology, 2007. 81(8): p. 3891-903. 44. Kaufmann, B. and M.G. Rossmann, Molecular mechanisms involved in the early

steps of flavivirus cell entry. Microbes and infection / Institut Pasteur, 2011. 13(1): p. 1-9.

45. Luo, D., et al., Crystal structure of the NS3 protease-helicase from dengue virus.

Journal of virology, 2008. 82(1): p. 173-83. 46. Navarro-Sanchez, E., P. Despres, and L. Cedillo-Barron, Innate immune

responses to dengue virus. Archives of medical research, 2005. 36(5): p. 425-35. 47. Morrison, J., S. Aguirre, and A. Fernandez-Sesma, Innate immunity evasion by

Dengue virus. Viruses, 2012. 4(3): p. 397-413. 48. Lund, J.M., et al., Recognition of single-stranded RNA viruses by Toll-like

receptor 7. Proceedings of the National Academy of Sciences of the United States of America, 2004. 101(15): p. 5598-603.

Page 40: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

29

49. Bukowski, J.F., et al., Dengue virus-specific cross-reactive CD8+ human cytotoxic T lymphocytes. Journal of virology, 1989. 63(12): p. 5086-91.

50. An, J., et al., Dengue-specific CD8+ T cells have both protective and pathogenic

roles in dengue virus infection. Immunology letters, 2004. 95(2): p. 167-74. 51. Kurane, I., A. Meager, and F.A. Ennis, Dengue virus-specific human T cell

clones. Serotype crossreactive proliferation, interferon gamma production, and cytotoxic activity. The Journal of experimental medicine, 1989. 170(3): p. 763-75.

52. Mangada, M.M. and A.L. Rothman, Altered cytokine responses of dengue-

specific CD4+ T cells to heterologous serotypes. Journal of immunology, 2005. 175(4): p. 2676-83.

53. Rothman, A.L., Immunity to dengue virus: a tale of original antigenic sin and

tropical cytokine storms. Nature reviews. Immunology, 2011. 11(8): p. 532-43. 54. Beltramello, M., et al., The human immune response to Dengue virus is

dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity. Cell host & microbe, 2010. 8(3): p. 271-83.

55. Valdes, K., et al., Human Dengue antibodies against structural and nonstructural

proteins. Clinical and diagnostic laboratory immunology, 2000. 7(5): p. 856-7. 56. Churdboonchart, V., et al., Antibodies against dengue viral proteins in primary

and secondary dengue hemorrhagic fever. The American journal of tropical medicine and hygiene, 1991. 44(5): p. 481-93.

57. Lei, H.Y., et al., Immunopathogenesis of dengue virus infection. Journal of

biomedical science, 2001. 8(5): p. 377-88. 58. Boonnak, K., et al., Role of dendritic cells in antibody-dependent enhancement of

dengue virus infection. Journal of virology, 2008. 82(8): p. 3939-51. 59. Kliks, S.C., et al., Antibody-dependent enhancement of dengue virus growth in

human monocytes as a risk factor for dengue hemorrhagic fever. The American journal of tropical medicine and hygiene, 1989. 40(4): p. 444-51.

60. Lozach, P.Y., et al., Dendritic cell-specific intercellular adhesion molecule 3-

grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals. The Journal of biological chemistry, 2005. 280(25): p. 23698-708.

61. Sabin, A.B., Research on dengue during World War II. American journal of

tropical medicine and hygiene, 1952. 1(1): p. 30-50.

Page 41: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

30

62. Schwartz, L.M., et al., The dengue vaccine pipeline: Implications for the future of

dengue control. Vaccine, 2015. 33(29): p. 3293-3298. 63. Coller, B.-A.G., et al., The development of recombinant subunit envelope-based

vaccines to protect against dengue virus induced disease. Vaccine, 2011. 29(42): p. 7267-7275.

64. Beckett, C.G., et al., Evaluation of a prototype dengue-1 DNA vaccine in a Phase

1 clinical trial. Vaccine, 2011. 29(5): p. 960-968. 65. ClinicalTrials.gov. Safety study of a dengue virus DNA vaccine. 2015 [cited

2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT00290147. 66. ClinicalTrials.gov. A two-dose primary vaccination study of a tetravalent dengue

virus purified inactivated vaccine vs. placebo in healthy adults. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01666652.

67. ClinicalTrials.gov. TDENV PIV and LAV dengue prime-boost strategy. 2014

[cited 2016 10-14-2016]; Available from: https://clinicaltrials.gov/show/NCT02239614.

68. Durbin, A.P., et al., rDEN4Δ30, a live attenuated dengue virus type 4 vaccine

candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers. Journal of Infectious Diseases, 2005. 191(5): p. 710-718.

69. Durbin, A.P., et al., Attenuation and immunogenicity in humans of a live dengue virus type-4 vaccine candidate with a 30 nucleotide deletion in its 3'-untranslated region. The American journal of tropical medicine and hygiene, 2001. 65(5): p. 405-413.

70. Wright, P.F., et al., Phase 1 trial of the dengue virus type 4 vaccine candidate

rDEN4Δ30-4995 in healthy adult volunteers. The American journal of tropical medicine and hygiene, 2009. 81(5): p. 834-841.

71. ClinicalTrials.gov. Evaluation of the safety and efficacy of a single dose of a

dengue vaccine (TV005) in healthy adults. 2015 [cited 2016 10-14-2016]; Available from: https://clinicaltrials.gov/show/NCT02317900.

72. ClinicalTrials.gov. Phase II trial to evaluate safety and immunogenicity of a

dengue 1,2,3,4 (attenuated) vaccine TV003/TV005. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01696422.

Page 42: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

31

73. Osorio, J.E., et al., Development of DENVax: a chimeric dengue-2 PDK-53-based tetravalent vaccine for protection against dengue fever. Vaccine, 2011. 29(42): p. 7251-7260.

74. ClinicalTrials.gov. Safety and immunogenicity study to assess DENVax, a live

attenuated tetravalent vaccine for prevention of dengue ever. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01224639.

75. ClinicalTrials.gov. Phase 1b study investigating safety & immunogenicity of

DENVax given intradermally by needle or needle free PharmaJet injector. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01765426.

76. ClinicalTrials.gov. Safety and immunogenicity of different schedules of Takeda's

tetravalent dengue vaccine candidate (TDV) in healthy participants. 2015 [cited 2016 10-14-2016]; Available from: https://clinicaltrials.gov/show/NCT02302066.

77. Guy, B., et al., From research to phase III: preclinical, industrial and clinical

development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine, 2011. 29(42): p. 7229-7241.

78. Sin Leo, Y., et al., Immunogenicity and safety of recombinant tetravalent dengue

vaccine (CYD-TDV) in individuals aged 2–45 years: Phase II randomized controlled trial in Singapore. Human vaccines & immunotherapeutics, 2012. 8(9): p. 1259-1271.

79. Amar-Singh, H., et al., Safety and immunogenicity of a tetravalent dengue

vaccine in healthy children aged 2–11 years in Malaysia: A randomized, placebo-controlled, Phase III study. Vaccine, 2013. 31(49): p. 5814-5821.

80. ClinicalTrails.gov. Study of ChimeriVax™ tetravalent dengue vaccine in healthy subjects. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT00875524.

81. ClinicalTrials.gov. Study of a novel tetravalent dengue vaccine in healthy children

aged 2 to 14 years in Asia. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01373281.

82. Guy, B. and N. Jackson, Dengue vaccine: hypotheses to understand CYD-TDV-

induced protection. Nature Reviews Microbiology, 2015.

Page 43: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

32

CHAPTER 2

BROAD NEUTRALIZING RESPONSES ELICITED BY FLAVIVIRUS

INFECTION IN CD46 INFarKO MICE

ABSTRACT Arthropod-borne flaviviruses are a very conservative group of viruses that share

the envelope glycoprotein protein structure that determines its rigid eicosahedral

symmetry. As such, antibodies directed against conserved regions of this envelope

glycoprotein lattice can precipitate cross-reaction, cross neutralization and cross

protection among diverse flavivirus depending on antibody concentration, specificity and

avidity. We showed here that the inoculation of CD46-IFNarko mice with one dengue

serotype could generate neutralizing antibodies against homologous and heterologous

dengue virus serotypes. This animal host is relevant because it will be use for subsequent

immunogenicity experiments. As such this chapter is relevant because it gives substrate

to the search of cross-neutralizing respnses elicited by vectored viruses. Animals were

inoculated with DV2 or DV4 elicited anti-DV1 antibodies at 28 days postinfection with a

titer of 1:140 and 1:160 PRNT50, respectively. Most interestingly, we show that serum

from animals inoculated with dengue 2 virus generates a more robust cross-neutralizing

immune response to Yellow fever virus when compared to serum from animals

inoculated with Dengue 4 virus: 1:853 and 1:20 PRNT50, respectively. Furthermore, in

this chapter, we documented the correlation between two different methods to quantify

neutralizing antibodies in serum, the classic plaque reduction neutralization titer method

at an end point of 50 (PRNT50) and the log neutralization index (LNI).

Page 44: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

33

IMPORTANCE

Dengue infections have been a major public heath concern. The data from the

recent clinical trial of CYD vaccine showed a new challenge for dengue vaccine

development. Naïve vaccinees with no previous exposure to any flavivirus infection were

at risk of generating severe dengue disease after receiving the tetravalent dengue vaccine

DengvaxiaMR. Here, we show that monotypic dengue infections generate a neutralizing

immune response to the homologous serotype as well as other dengue serotypes and

yellow fever. The titers of the neutralizing antibodies were different based on how closely

related the tested virus is. This finding is important because supports the notion of cross-

neutralizing protection given an adequate antigenic stimulus for vaccination.

Page 45: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

34

INTRODUCTION Clinical responses to dengue infections are determined by the previous flaviviral

exposure; individuals who experienced primary dengue infection are at high risk in

generating a severe dengue disease upon a secondary, heterologous dengue infection than

dengue naïve people. Therefore, tetravalency has been a sine-equa-non requirement to in

dengue vaccine development to prevent the enhancement of secondary dengue

infection[84]. In monotypic dengue infections, young children are 6–7 times more likely

to be hospitalized with a secondary DV infection than are adults[85]. Furthermore, the

sequence of DV infection determined the clinical outcomes of the secondary infection.

For instance infections in the series DV1 then DV 2 or DV 3 are more likely to be severe

in nature than DV 2 then DV 3[86-88]. The enhancement of dengue disease in secondary

infection is explained by the presence of neutralizing antibodies at sub-protective levels.

However, such an effect does not occur in flavivirus seropositive patients. Patients with

Japanese encephalitis virus (JEV) immunity, either due to prior wild-type JEV infection

or immunization with the corresponding attenuated or inactivated vaccine, may

experience dengue infection that range in outcome from in apparent to mild disease, but

very rarely progress to severe forms of dengue disease[89]. The same observation is

verified among multitype DV immune individuals, where most of third DV infections

result in mild-moderate febrile disease that rarely requires hospitalization depite the fact

of the presence of antibodies against three DV serotypes[90]. The evaluation of clinical

responses to Sanofi Pasteur dengue vaccine in naïve, younger than five years of age

recipients, provided evidence for vaccine enhanced dengue disease, which is dependent

Page 46: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

35

upon the serological status of vaccine recipients[83, 91]. Despite the presence of

neutralizing antibodies against the four DV serotypes, this group of vaccinees

experienced an enhancement of dengue disease. In seronegative children of all ages,

severe forms of dengue disease occurred more often in vaccinated (18/65) than in non-

vaccinated groups (6/39). In the Asian phase III trial (CYD14), vaccine efficacy in

seropositive children was somewhat lower in <9-year-old than ≥9-year-old seropositive,

70.1%, and 79.2%, respectively. However, in seronegative recipients, vaccine efficacy

was modest, in <9 year-olds as low as 14.4%. As a result, vaccinated seronegative

individuals presumably will retain risk to enhanced DV disease for many years[92, 93].

Therefore, it is imperative to consider improving the recipient’s serological status in

order to complement the use of the Sanofi vaccine to prevent severe forms of DV disease.

In this chapter, we explore viral neutralization among flaviviruses in CD46-

IFNarko mice immunized with DV2 or DV4. We demonstrated that dengue infection

could generate cross-neutralizing antibodies responses to heterologous dengue serotypes

(DV1) as well as relatively distance flavivirus (YFV 17). In addition we showed the

correlation between two different methods to quantify neutralizing antibodies: the classic

method, PRNT50, dependent on serial dillutions of tested serum to measure the dilution

that causes a 50% reduction in a determined number of plaques, and the LNI method that

depends on mixing equal volume of the virus with serum and thus measures serum

impact on viral infectivity. We observed that LNI gaves us a larger dynamic range of

neutralization measurements, furthermore LNI represents the only vaccine correlate of

protection for a flavivirus, YFV.

Page 47: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

36

MATERIALS & METHODS

Cells and viruses.

BHK 21 cells were maintained in DMEM (DMEM, Sigma-Aldrich, St. Louis,

MO, USA) supplemented with 10% FBS (FBS, Atlanta Biologicals, Flowery Branch,

GA, USA), and 1% penicillin-streptomycin (Sigma-Aldrich). DV1, DV2, and DV4

viruses were obtained from the ATCC (VR-1586 strain TH Soman, VR-1584 strain New

Guinea C and VR-1257 strain H241, respectively) and propagated in Vero cells. The

viruses were then amplified in BHK21 cells for up to two or three passages. YFV 17D

viral strain was obtained using reverse genetic techniques in BHK-21 cells from the full-

length plasmid pACNR-FLYF\17 [94]and propagated in a similar way as DV.

Mice inoculations.

All experimental procedures were performed according to a protocol approved by

the Arizona State University Institutional Animal Care and Use Committee. Mice were

inoculated by the intraperitoneal (i.p.) route with the indicated doses of 10^5 PFU/ml

diluted in 100 uL of Opti-MEM. Five weeks after the inoculation, mice were

exsanguinated. Sera were separated and heat-inactivated at 56°C for 1 h[95].

DV and YFV neutralization assay.

To determine DV2, DV4, DV1, and YFV Log10 Neutralization Index titer the

corresponding viral prep was mixed with heat-inactivated serum in a 1:1 proportion in a

25 uL volume of Opti-MEM and incubated for 1 h at 37 °C. Infectivity after serum

incubation was determined using a plaque assay in BHK-21 cells. Briefly, ten-fold serial

dilutions of the virus/serum mixtures were generated using Opti-MEM and added to 2.4 ×

Page 48: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

37

106 BHK-21 cells in a 24-well plate and then incubated for 4 h at 37 °C. At this time, 500

uL of 3% carboxymethyl cellulose was added to each well and plates were incubated for

5 days at 37 °C. The wells were then washed; the monolayer was fixed and stained with

acetic acid/naphtol blue-black and plaques were counted. The neutralization index was

expressed as the logarithmic difference between the averaged DV2, DV4, DV1 or YFV

titers when incubated with control, namely, that of animals inoculated with MVvac2, and

preimmune. Neutralization indexes thus determined were graphed as Log10NI.

In another approach, we performed a standard 50% plaque reduction

neutralization assay (PRNT50) using pooled sera from each experimental group. Briefly,

50 DV2, DV4, DV1 or YFV PFU were incubated with two-fold serum dilutions (from

1:5 to 1:320) for 1 h at 37 °C. Remaining infectivity was determined by plaque assay, as

described above. The neutralization titer was defined as the dilution that resulted in a

50% reduction in plaque numbers, as compared to unrelated serum.

Correlation of PRNT50 and LNI

Serum obtained from single animal inoculated by either DV2 or DV4 was used in

this experiment. Two fold serum dilution (from 1:5 to 1: 12560) was incubated with 50

PFU of DV2 or DV4 to perform a standard 50% plaque reduction neutralization assay

(PRNT50), as described above. The same dilution was used to perform a Log10

Neutralization Index titer by mixing the corresponding virus in a ration of 1:1 to the

diluted serum in 25 ul volume and incubated at 37C for 1 hr. Infectivity after serum

incubation was determined as described above.

Page 49: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

38

RESULTS

Monotypic Dengue virus immunity

To rank the immunogenicity of homologous dengue serotypes, we immunized

groups of three huCD46Ge-IFNarKO mice with one 105 PFU/ml intraperitoneal dose of

DV2 or DV4, and analyze neutralizing immune responses siw weeks postinoculation. We

assessed DV2 or DV4 neutralization using two related approaches. First, a Log10

neutralization index (LNI), where the neutralizing effects of individual 1:2 serum

dilutions upon a high titer viral DV2 or DV4 prep infectivity was calculated using

averaged preimmune measurements to determine basal, non-neutralizing conditions. Thus

preimmune, or non-reactive samples are set for an LNI of zero. Additionally, we

determined the classic PRNT50 titer. As shown in figure 2.1A, serum samples from a

group of animals inoculated by DV2 reduced the infectivity of a 105.48 PFU/ml DV2 viral

prep by 4.2 orders of magnitude after one dose, which is equivalent to a PRNT50 of

1:1280. In figure 2.1C, serum samples from a group of animals inoculated by DV4

reduced the infectivity of DV4 (106 PFU/ml) by 4.63 orders of magnitude after one dose,

which is equivalent to PRNT50 of 1: 2133.3.

Subsequently, we explored the correlation between the titer of neutralizing anti-

DV2 or DV4 expressed by LNI and PRNT50. As shown in figure 2.1B and 2.1D, there is

a consistent correlation between the titer of neutralizing antibodies expressed either by

LNI or PRNT50.

Page 50: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

39

Heterologous cross-neutralization immunity to DV1 and YFV

To explore the heterologous immune response of DV2 and DV4 we performed

viral neutralization assays against DV1 and the more phylogenetically distant YFV 17D.

Figure 2.2 documents that serum samples from four groups of the experimental animals

received one dose of DV1, DV2, DV4 and YFV measurably neutralize these two

flaviviruses. The infectivity of a high titer (104.84 PFU/ml) DV1 viral prep was

neutralized for an averaged LNI of 4.2 equivalent to 1:960 PFU after one dose DV1

vaccination. A lower titer was obtained after DV2 or DV4 one-dose inoculation scheme.

The documented DV1 LNI neutralization titer for DV2 and DV4 experimental groups

was 2.2 and 1.9, respectively, which equivalent to 1:426 and 1: 480 PRNT50 (Fig 2.2A).

In a comparable result, all of the animals neutralized YFV. An YFV 17D high

titer viral prep (106.78 PFU/ml) infectivity was neutralized by an LNI average of 3.86 and

0.9) equivalent to a PRNT50 of 1:853 and 1:20 after DV2 or DV4 inoculation,

respectively. Of note, an LNI of 0.7 is the reported correlate of protection for YFV

(equivalent approximately to a PRNT50 of 1:10)[96]. The YFV LNI neutralization titer

of YFV experimental group was 4.13, which equal to 1:1706 PRNT50 (Figure2.2B).

Page 51: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

40

DISCUSSION

In this chapter, we have shown that immunization by DV2 or DV4 can elicit

neutralizing immune responses in protective ranges against the homologous DV2, or

DV4 and also against the heterologous DV1 and the relatively phylogenetically distant

YFV 17D attenuated strain. It has been known that primary infection with one-dengue

serotypes can generate a long-term protection against that serotype and a short term

(around one month) protection against any of the other three DV serotypes[97]. The

efficacy of a tetravalent dengue vaccine depends on the presence of flavivirus

immunity[91, 93]. Therefore, immunization strategy using a cocktail of different

flavivirus can be used to protect against a wide range of flavivirus infection including the

four dengue serotypes[98]. There are two important factors when we consider cross

protection against flaviviruses, the level of the cross-neutralizing antibodies, and the type

of flaviviruses that circulate in that endemic area[99]. Human volunteers inoculated seven

years previously with 17D yellow fever responded with a rapid production of neutralizing

antibodies to yellow fever and dengue type 1 and 2 when inoculated with an attenuated

DV2. The ability of these individuals to respond with anamnestic neutralizing antibodies

when infected with an heterologous flavivirus obeys to their previous exposure to

relatively closely flavivirus due to conserved artgropod-borne flaviral structures[100].

Such a concept is very critical when considering the development of dengue vaccine, as it

can be used as a novel strategy to design a vaccination strategy using a cocktail of closely

related flaviviruses with a combination of different serotypes of dengue. Multiple

observations of cross protection among flavivirus using different animal hosts have been

Page 52: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

41

reported. Sequential immunization of spider monkeys with 17D YFV, DV2, and JEV

protected animals from challenges by DV1, 3, and 4[99]. Inoculation of hamsters with

DV1 or DV2 protect against a West Nile (WN) virus or St. Louis encephalitis virus

challenge[101]. In a remarkable work by Winston Price, spider monkeys immunized

sequential with 17D YFV, live attenuated Langat E5 virus, and live attenuated DV2 were

protected against viral challenges with more than 14 different flavivirus including

DV1,2,3,4, JEV, WNV, and Zika. Importantly, this sequential immunization did not only

protect monkeys against all four dengue serotypes but also generated a solid immunity

against the four serotypes as determined by no dengue antibody booster HI responses,

which supports the notion that the sequential immunization with 3 related flaviviruses

would not cause enhancement of dengue disease[98]. This strategy depends on the

sequence of the immunization and type f flavivirus election; the virus chosen for the use

in vaccination must give the broadest heterologous protection[102].

In sum, we showed that immunization with type 4 or 2 dengue virus generates

cross-neutralizing immune responses to 17D YFV. Unexpectedly, animals inoculated by

DV2 had a more robust neutralizing response to YFV than animals immunized by DV4,

despite the fact that DV4 is more closely related to YFV phylogenetically. More work is

needed to test the perfect combination of viruses to generate the broadest neutralizing

immune response among different flaviviruses. In the following chapter, we documented

the homologous immune responses and heterologous cross immune response of animals

inoculated by recombinant measles expressing monovalent dengue antigens or a cocktail

of recombinant measles expressing divalent dengue antigens.

Page 53: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

42

Figure2.1. Monotypic immunogenicity after Dengue 2 and 4 inoculation. Mice received one IP dose of DV2 or DV4. Neutralizing immune responses were analyzed 32 days later. (A) and (B) Analysis of neutralization against Dengue 2; and (C) and (D) against Dengue 4 viruses. Anti-dengue neutralizing immune responses are assessed by LNI (left panel) and PRNT50 (right panel) in A and C. Each symbol represents one animal; horizontal lines and vertical brackets indicate group average and standard deviation, respectively. In B and D the correlation between anti-dengue LNI (Y axes) and PRNT50 (X axes) were determined.

Page 54: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

43

Figure 2.2. Cross-neutralizing immune responses in mice inoculated with DV1, DV2, DV4, and YFV (A) Analysis of neutralization against Dengue 1 and (B) against attenuated YFV. Anti-dengue and anti-YFV neutralizing immune responses are assessed by LNI (left panel) and PRNT50 (right panel). Averages of anti-Dengue and Anti-YFV LNI and PRNT50 titers determined in triplicate of pooled serum samples are indicated.

Page 55: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

44

REFERENCES

1. Wilder-Smith, A. and D.J. Gubler, Dengue vaccines at a crossroad. Science, 2015. 350(6261): p. 626-627.

2. Guzman, M.a.G., et al., Effect of age on outcome of secondary dengue 2

infections. International journal of infectious diseases, 2002. 6(2): p. 118-124. 3. Guzman, M.G., et al., Epidemiologic studies on Dengue in Santiago de Cuba,

1997. American journal of epidemiology, 2000. 152(9): p. 793-799. 4. Guzman, M.G., et al., Epidemiological studies on dengue virus type 3 in Playa

municipality, Havana, Cuba, 2001–2002. International journal of infectious diseases, 2012. 16(3): p. e198-e203.

5. OhAinle, M., et al., Dynamics of dengue disease severity determined by the

interplay between viral genetics and serotype-specific immunity. Science translational medicine, 2011. 3(114): p. 114ra128-114ra128.

6. Anderson, K.B., et al., Preexisting Japanese encephalitis virus neutralizing

antibodies and increased symptomatic dengue illness in a school-based cohort in Thailand. PLoS Negl Trop Dis, 2011. 5(10): p. e1311.

7. Kosasih, H., et al., Report of four volunteers with primary, secondary and tertiary

dengue infections during a prospective cohort study. Dengue Bulletin, 2006. 30: p. 87.

8. Capeding, M.R., et al., Clinical efficacy and safety of a novel tetravalent dengue

vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. The Lancet, 2014. 384(9951): p. 1358-1365.

9. Guy, B. and N. Jackson, Dengue vaccine: hypotheses to understand CYD-TDV-

induced protection. Nature Reviews Microbiology, 2015. 10. Hadinegoro, S.R., et al., Efficacy and long-term safety of a dengue vaccine in

regions of endemic disease. New England Journal of Medicine, 2015. 373(13): p. 1195-1206.

11. Villar, L., et al., Efficacy of a tetravalent dengue vaccine in children in Latin

America. New England Journal of Medicine, 2015. 372(2): p. 113-123. 12. Bredenbeek, P.J., et al., A stable full-length yellow fever virus cDNA clone and

the role of conserved RNA elements in flavivirus replication. Journal of general virology, 2003. 84(5): p. 1261-1268.

Page 56: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

45

13. Mrkic, B., et al., Measles virus spread and pathogenesis in genetically modified

mice. Journal of virology, 1998. 72(9): p. 7420-7427. 14. Mason, R.A., et al., Yellow fever vaccine: direct challenge of monkeys given

graded doses of 17D vaccine. Applied microbiology, 1973. 25(4): p. 539-544. 15. SANGKAWIBHA, N., et al., Risk factors in dengue shock syndrome: a

prospective epidemiologic study in Rayong, Thailand I. The 1980 outbreak. American journal of epidemiology, 1984. 120(5): p. 653-669.

16. Price, W.H., et al., Sequential immunization procedure against group B

arboviruses using living attenuated 17D yellow fever virus, living attenuated Langat E5 virus, and living attenuated dengue 2 virus (New Guinea C isolate). The American journal of tropical medicine and hygiene, 1973. 22(4): p. 509-523.

17. PRICE, W.H., Sequential immunization as a vaccination procedure against

dengue viruses. American journal of epidemiology, 1968. 88(3): p. 392-397. 18. Schlesinger, R.W., et al., Clinical and serologic response of man to immunization

with attenuated dengue and yellow fever viruses. The Journal of Immunology, 1956. 77(5): p. 352-364.

19. PRICE, W.H. and I.S. THIND, Protection against West Nile virus induced by a

previous injection with dengue virus. American journal of epidemiology, 1971. 94(6): p. 596-607.

20. Price, W.H., et al., A sequential immunization procedure against certain group B

arboviruses. The American journal of tropical medicine and hygiene, 1963. 12(4): p. 624-638.

Page 57: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

46

CHAPTER 3

GENERATION OF A COMPLEMENTARY ANTI-DENGUE IMMUNIZATION

STRATEGY BASED ON A RECOMBINANT MEASLES VIRUS VACCINE

COCKTAIL

ABSTRACT

Despite the approval of a Dengue virus (DV) vaccine in five endemic countries,

dengue prevention would benefit from an immunization strategy highly immunogenic in

young infants and not curtailed by viral interference. Vaccine efficiency is majorly

improved when the recipient’ immune system has been previously primed against related

flaviviruses. To create a pediatric vaccination strategy with the potential to prime

neutralizing anti-flaviviral responses in one-year-old infants, we modified a measles

vaccine viral vector (MVvac2) to express the pre membrane (prM) and a truncated (90%)

form of the major envelope (E) from DV2 and DV4. Single-component recombinant MV

vaccines elicited robust homologous neutralizing responses in all MV-susceptible mice

(1:214 and 1:640 PRNT50 against DV2 and DV4 in average). When MV-DV 2/4

recombinant vaccines were combined, we documented an enhancement in homologous

neutralizing responses and cross-neutralizing immune responses against DV 1 and YFV

17D in all experimental animals. Thus, we can prime broad neutralizing immune

responses using only two of the four available DV serotypes. Using the current MV

immunization scheme to prime anti-flaviviral neutralizing immunity in young infants

Page 58: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

47

may be a highly convenient and cost-effective strategy to robustly enhance the

immunogenicity and efficiency of subsequent chimeric Dengue vaccine approaches.

Page 59: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

48

IMPORTANCE

The four dengue virus serotypes are the cause of the most important arthropod-

borne viral disease worldwide. After six decades of research, a dengue vaccine has been

approved in five countries and another two candidates are approximating approval. The

two most important issues documented so far are the lack of efficiency in young

recipients and an uneven protection against the four DV serotypes. Since the dengue

vaccine performs better when the host has previous anti-flaviviral immunity, we proposed

here an attractive solution to both issues by using a recombinant measles vaccine

vectoring dengue protective antigens to be administered to young infants. We have

generated recombinant measles virus expressing Dengue 2 and 4 antigens that

successfully induce neutralizing responses in experimental animals. Interestingly, when

combined, the vaccine cocktail is able to prime cross-neutralizing responses against DV 1

and the relatively distant 17D yellow fever virus attenuated strain. The vaccine

equivalency of our MVvac2 viral vector suggests usage of a recombinant measles-dengue

cocktail immunization to prime a broad, anti-flaviviral response in one year-old recipients

that would be majorly boosted when the chimeric Dengue vaccine approach is used later.

Page 60: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

49

INTRODUCTION

In terms of human suffering and economic impact, infections caused by any of the

four existing Dengue virus (DV) serotypes cause the most important arthropod-borne

viral infectious disease in the world. Currently, it is estimated that almost half of the

globe population is at risk of contracting Dengue, mainly people living in Asia, Africa

and Latin America, in this order. With an annual incidence of Dengue clinical cases

estimated at 100 million and a case fatality rate of 1%, the introduction of a vaccine

strategy for countries with a high prevalence of dengue has been an important advance to

control this devastating infectious disease[103, 104].

As for other arthropod-borne flaviviruses such as Yellow Fever virus (YFV)

Japanese encephalitis virus or Thick-borne encephalitis virus, protective immunity

against DV is associated with the development of neutralizing antibodies[105]. These are

majorly directed against the envelope (E) glycoprotein[106, 107]. DV envelope displays

an icosahedral array of ninety E dimers held structurally in place by a subjacent

membrane (M) protein network[35]. After a natural DV infection, a serotype-specific and

long-lived neutralizing immunity is developed, thus the formulation of a tetravalent

vaccine with the capability of eliciting viral neutralization has been the goal for more

than six decades[55, 63]. The currently approved DV vaccine (DengvaxiaTM) consist of a

tetravalent formulation of attenuated chimeric viruses where the envelope glycoproteins

from the relevant DV serotype enclose the replication machinery of the attenuated YFV

17D vaccine strain. After phase three clinical trials and a careful analysis of

immunogenicity and vaccine efficiency, Dengvaxia was approved for use in highly

Page 61: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

50

dengue endemic countries targeting 9-45 years-old population, exclusively. This

indication represents the main limitation for this vaccination strategy, namely an

individual with a high probability of having pre-existent immunity against Dengue [91,

108-111].

In contrast, measles virus (MV) vaccination has been available for more than six

decades with excellent records of safety and efficiency. The current two-dose vaccine

schedule recommended by the WHO is credited to be responsible of eliminating

indigenous MV transmission in the American continent and is the spearheading strategic

effort to eradicate this infectious disease[112].Ours and other groups have proposed to

use viral vectors with MV vaccine equivalency to generate divalent and multivalent

vaccine options[14-16]. To give substrate to this overarching aim we have documented

that recombinant MV vaccines remain protective against a measles challenge [113].

In this paper, we showed that vectored MV-DV2 and MV-DV4 vaccines expressing

relevant DV envelope proteins and administered simultaneously, are able to elicit broad

neutralizing immune responses in MV-susceptible mice. Using MV vaccination to

purposefully enhance DV seroprevalence in the very young has the potential to expand

the indications of the current DV vaccine.

Page 62: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

51

MATERIALS AND METHODS

Cells and Viruses

Vero/hSLAM cells[114, 115] were maintained in Dulbecco’s modified Eagle’s

medium with high glucose concentration (DMEM, Sigma-Aldrich, St. Louis, MO, USA)

supplemented with 5% fetal bovine serum (FBS, Atlanta Biologicals, Flowery Branch,

GA, USA), 1% penicillin–streptomycin (Sigma-Aldrich) and 0.5 mg/mL G418 (Enzo

Life Sciences, Farmingdale, NY, USA) to maintain hSLAM expression selection. BHK

21 cells were maintained in DMEM supplemented with 10% FBS and 1% penicillin-

streptomycin. Helper 293-3-46 cells [116]a derivative from HEK 293 cells were

maintained in DMEM with 10% FBS, 1% PS, and 1.2 mg/mL G418.

DV1, DV2 and DV4 viruses were obtained from the ATCC (VR-1586 strain TH

Soman, VR-1584 strain New Guinea C and VR-1257 strain H241, respectively) and

propagated in Vero cells. The viruses were then amplified in BHK21 cells for up to two

or three passages. YFV 17D viral strain was obtained using reverse genetic techniques in

BHK-21 cells from the full length-plasmid pACNR-FLYF\17[94] and propagated in a

similar way as DV.

Recombinant MVs were generated using the method of Radecke et al. [117]

modified by Parks et al.[118]. After detection of their cytopathic effect on mixed cultures

of Helper 293-3-46 and Vero/hSLAM cells, individual syncytia were transferred to and

propagated in Vero/hSLAM cells. Viral clones stocks were amplified by infecting

Vero/hSLAM cells at a multiplicity of infection (MOI) of 0.03 and incubated at 37°C.

When approximately 80% cytopathic effect was observed, cells were scraped in Opti-

Page 63: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

52

MEM (Life Technologies, Grand Island, NY) and viral particles were released by two

freeze-thaw cycles.

Multi-step growth kinetics of the viruses were determined by infecting

Vero/hSLAM cells at an MOI of 0.03 and incubating them at 37°C. Infected cells and

supernatants were collected and lysed by a single cycle of freeze-thaw at prescribed times

post-infection, and the 50% tissue culture infectious dose (TCID50) was determined in

Vero/hSLAM using the Spearman-Kärber end-point dilution method [119]. Primary

chick embryo fibroblast (CEF) were obtained from ATCC (CRL-1590 F2) and grown in

Dulbecco’s modified Eagle’s medium with high glucose concentration (DMEM, Sigma-

Aldrich, St. Louis, MO, USA) supplemented with 10% FBS, 1% penicillin–streptomycin

and 10% tryptose phosphate broth (Gibco, Life Technologies). Kinetics of infection of

CEF were performed by infecting CEF at MOI of 0.03 with MV and DV-expressing

recombinant MV and collecting cell-associated and supernatant at the indicated time

post-infection.

Plasmids and plasmids constructions

The plasmid pB(+)MVvac2(HBsAg)N was used to construct the recombinant

MVs expressing DV antigens. This plasmid contains an infectious MV cDNA

corresponding to the antigenome of the Schwarz measles vaccine strain [113, 120] with

an additional transcription unit (ATU) inserted downstream of the nucleocapsid (N)

cistron to direct the expression of an inserted foreign gene. To generate the recombinant

measles expressing DV2 or DV4 prM- E or prM-Esol glycoproteins, MluI and AatII

digestion fragments containing the desired coding sequence were obtained by PCR

Page 64: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

53

(primer sequence available upon request) and swapped with the HBsAg insert. DV2

prME coding sequences were obtained from the full-length PD2/I/C/30P/A plasmid

(strain 16681[121, 122], and DV4 prME coding sequences were obtained from the full-

length pR424-D4 plasmid (H241 strain). The recombinant full-length plasmids were

termed pB(+)MVvac2-DV2(prME)N, pB(+)MVvac2-DV2(prMEsol)N, pB(+)MVvac2-

DV4(prME)N and pB(+)MVvac2-DV4(prME)N. Correct insertions and relevant gene

boundaries were corroborated by sequencing.

Indirect Immunofluorescence Microscopy

Vero/hSLAM cells were seeded into 4 well glass Millicell EZ slides

(MILLPORE) and infected at MOI of 0.06, 0.1 and 1 with MV, recombinant MV,

DENV2 and 4, respectively. After 48 hours of incubation at 37 C, the cells were fixed

with 3% paraformaldehyde and incubated at 37 °C overnight. The cells were

permeabilized with 0.1% Triton X-100 in phosphate- buffer saline (PBS). Both primary

and secondary antibodies dilution were prepared in 0.1% triton X-100 in 10% FBS serum

in 1 X PBS. Two Mixes of primary antibodies were used, monoclonal antibody directed

against DENV 1,2,3,4 E protein at 1:50 dilution (a mouse IgG2a cat. # 76 ab9202 from

abcam) with the polyclonal antibodies directed against MV N protein at 1:300 dilution (a

rabbit IgG antibody provided from R. Cattaneo). The primary antibodies incubations

were carried out overnight at 4 °C. A mix of goat anti-mouse IgG coupled to Fitc (Green

fluorescence), and a goat anti-rabbit IgG coupled to PE (Red fluorescence) were used as a

secondary antibody with dilution of 1:300 and incubated at room temperature for 2 hours.

The coverslips were mounted on slides by using flouromount G with DAPI (Electron

Page 65: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

54

Microscopy Science). The slides examined and images were taken by inverted

microscope with epifluorescence illumination (EVOSFL).

Western Blot Assay

Vero/hSLAM cells were seeded in 100-mm-diameter dishes and infected at an

MOI of 0.3 with MV or recombinant MVs, or were mock-infected. Forty h after

infection, cells were washed three times with phosphate-buffered saline (PBS) then lysed

with RSB-NP40 buffer (1.5 mM MgCl2, 10 mM Tris-HCl, 10 mM NaCl, and 1%

Nonidet P-40, Sigma-Aldrich, St. Louis, MO) plus protease inhibitors (cOmplete Mini

Protease Inhibitor Tablets, Roche Diagnostics, Manheim, Germany). The protein extracts

were mixed with Laemmli buffer (Bio-Rad Laboratories, Hercules, CA) containing β

mercaptoethanol and denatured at 96C.

25 ul of protein extraction loaded and separated by polyacrylamide gel

electrophoresis (PAGE) in a 4-15% or 10% acrylamide gel. Following electrophoretic

separation, proteins were transferred to nitrocellulose for immunoblotting using a

1:20,000 dilution of rabbit polyclonal anti-MV N, a 1:100 dilution of a mouse

monoclonal antibody directed against DV2 E DIII (GTX29202, GeneTex, Irvine, CA,

USA) or 1:100 dilution of a mouse monoclonal antibody directed against DV4 E protein

(clone E62, VR-1726, ATCC, VA, USA) , or a 1:10,000 dilution of mouse anti-actin

(Sigma-181 Aldrich, St. Louis, MO), followed by the appropriate horseradish peroxidase

(HRP) conjugated secondary antibody (GE Healthcare, Little Chalfont, United

Kingdom). Reactions were developed using a chemiluminescence kit (SuperSignal West

Pico Chemiluminescent Substrate, Pierce Biotechnology, Rockford, IL, USA).

Page 66: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

55

Glycosidase treatment.

MVvac2DV2(PrME)N or MVvac2DV2(PrMEsol)N infected cell lysates were

treated with endoglycosidase H (endo-H) and peptide N-glycosidase F (PNGase-F) (both

from NEB, Ipswich, MA) as per the manufacturer’s instructions for 1 h at 37°C. Protein

material was analyzed by immunoblotting using anti-H- or anti-E-specific antibodies.

Mice inoculations.

All experimental procedures were performed according to a protocol approved by

the Arizona State University Institutional Animal Care and Use Committee. To determine

the immunogenicity of recombinant MVs in a MV susceptible small animal host, groups

of HuCD46Ge-IFNarKO mice [123] were inoculated by the intraperitoneal (i.p.) route

with the indicated doses diluted in 100 uL of Opti-MEM. Three weeks after the initial

inoculation, the mice received a similar booster dose. Two weeks after the second dose,

mice were exsanguinated. Sera were separated and heat-inactivated at 56°C for 1 h.

MV neutralization assay.

To determine MV neutralization titer, serial two-fold dilutions of heat-inactivated

sera in Opti-MEM were incubated with 100 TCID50 of MVvac2 for 1 h at 37 °C.

Vero/hSLAM cells were added to the serum–virus mixtures and the plates were then

incubated at 37 °C for three days, at which time neutralization titer was assessed as the

highest dilution of serum capable of complete neutralization of infectivity, defined as the

Page 67: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

56

absence of MV cytopathic effect[124]. Neutralization titers were reported as the average

of a determination made in triplicate.

DV and YFV neutralization assay.

To determine DV2, DV4, DV1, and YFV Log10 Neutralization Index titer the

corresponding viral prep was mixed with heat-inactivated serum in a 1:1 proportion in a

25 uL volume of Opti-MEM and incubated for 1 h at 37 °C. Infectivity after serum

incubation was determined using a plaque assay in BHK-21 cells. Briefly, ten-fold serial

dilutions of the virus/serum mixtures were generated using Opti-MEM and added to 2.4 ×

106 BHK-21 cells in a 24-well plate and then incubated for 4 h at 37 °C. At this time, 500

uL of 3% carboxymethyl cellulose was added to each well and plates were incubated for

5 days at 37 °C. The wells were then washed, the monolayer was fixed and stained with

acetic acid/naphtol blue-black and plaques were counted. The neutralization index was

expressed as the logarithmic difference between the averaged DV2, DV4, DV1 or YFV

titers when incubated with control, namely, that of animals inoculated with MVvac2, and

preimmune. Neutralization indexes thus determined were graphed as Log10NI. In another

approach, we performed a standard 50% plaque reduction neutralization assay (PRNT50)

using pooled sera from each experimental group. Briefly, 50 DV2, DV4, DV1 or YFV

PFU were incubated with two-fold serum dilutions (from 1:5 to 1:320) for 1 h at 37 °C.

Remaining infectivity was determined by plaque assay, as described above. The

neutralization titer was defined as the dilution that resulted in a 50% reduction in plaque

numbers, as compared to unrelated serum.

Page 68: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

57

RESULTS

Generation and characterization of recombinant MVs expressing Dengue 2 envelope

antigens

Initially, to generate a MV-DV2 divalent vaccine vector, we modified a full-

length infectious cDNA plasmid named p(B(+)MVvac2. This plasmid and its derivatives

can be used to recover recombinant MVs by well-established reverse genetic techniques

[125, 126]. Two forms DV2 prME coding sequences were inserted downstream MV N

gene. The first one consisted of prM and E full-length coding sequences preceded by the

signal sequence of the carboxyl terminus of the capsid protein (C anchor). The recovered

virus was termed MVvac2-DV2(prME)N and expresses intact full length prM-E

glycoproteins. The second insertion consisted of similar C-anch, prM and a truncated

version of DV2 E. We deleted 45 amino acids from its carboxyl terminus corresponding

to E cell membrane anchor and cytoplasmic tail. We named the virus derived from this

construction MVvac2-DV2(prMEsol)N because this virus expresses an unattached

version of the DV2 E glycoprotein (Fig 3.1A). To corroborate the effect of DV2 prME

insertions upon recombinant MV replication fitness, we performed multistep growth

analysis using two cell substrates. Initially, viral replication in Vero/hSLAM did not

demonstrate a major deleterious effect because of the genetic modification on MV

backbone. Intracellular MVvac2-DV2(prME)N viral yields reached 6.6 Log10 TCID50/ml

at 48h PI, which were slightly higher than but not significantly different of the parental

strain MVvac2. MVvac2-DV2(prMEsol)N had a measurable negative effect on viral

replication in Vero/hSLAM cells, reaching intracellular titers of 5.8 Log10 TCID50/ml at

Page 69: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

58

the same time point and a slightly delayed viral growth kinetics. The extracellular

fractions for both viruses had similar patterns of replication with viral yields diminished

by one log10 approximately in comparison to the intracellular fraction (Fig3.1B). Since

both recombinants are attenuated MV strain (Moraten/Schwarz) derivatives, we analyzed

their replication in a cell line used for commercial production, primary chick embryo

fibroblasts. In these cells we observed a very similar replication profile for both

recombinants when compared to the parental MVvac2, reaching maximum intracellular

titers of around 5 log10 TCID50/ml at 6 days PI and extracellular titers of 102.5 TCID50/ml

(Fig 3.1C). Furthermore, plaque morphology analysis showed that MVvac2-

DV2(prME)N and MVvac2-DV2(prMEsol)N had circular plaques of around 1.075 and

1.045 mm in diameter respectively and not majorly different from MVvac2 ones (1.078

mm, Fig 3.1D).

Expression of Dengue 2 E glycoprotein by recombinant viruses

To corroborate the expression of DV2 vectored antigens, we performed initially

an indirect immunofluorescence assays using antibodies specific for MV-N (stained with

secondary Rhodamine conjugated antibodies), DV2 prM and E glycoproteins (stained

with FITC conjugated antibodies). To track the viral cytopathic effect, cell nuclei were

counter stained with DAPI. As evident in Figure 3.2A, merged epifluorescence images

from cells infected with either MVvac2-DV2(prME)N or MVvac2-DV2(prMEsol)N had

a strong yellow epifluorescence resulting from co-localization of green and red signals

coming from bona-fide cell syncitia induced by viral infection. In contrast control cells

infected with MVvac2 or DV-2 and treated in parallel similarly, showed only red and

Page 70: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

59

green epifluorescence, respectively. A similar result was obtained with a specific anti-

prM antibody. Thus, MVvac2-DV2(prME)N and MVvac2-DV2(prMEsol)N express the

expected DV-2 glycoproteins and the insertion of full-length or 90% DV2 prME coding

sequences did not have a major effect on MV replication in two different cell substrates.

Then, we analyzed biochemically the vectored DV2 E antigen. As shown in the left panel

of Figure 3.2B a specific monoclonal anti-DV2 E antibody detected protein bands of 52

and 48 kDa in lysates obtained from cells infected with MVvac2-DV2(prME)N and

MVvac2-DV2(prMEsol)N respectively. The approximately 4 kDa difference in the

vectored DV2 E molecular weight certainly obeys to the 45 amino acid carboxyl terminus

truncation of E as expressed by MVvac2-DV2(prMEsol)N. To demonstrate that such

modification of DV2 E promote its routing towards cell secretion, an analysis with

EndoH and PNGaseF glycosidases was performed. The right panel of Figure 3.2C

showed that and endoH-resistant DV2E form was detected in treated lysates from cells

infected with MVvac2-DV2(prMEsol)N in a clear contrast with MVvac2-DV2(prME)N

infected cell lysates treated similarly. To control this experiment, the same infected cell

lysates were probed with an anti-H antibody. Endo-H resistant MV H were detected in

both MVvac2-DV2(prME)N and MVvac2-DV2(prMEsol)N infected cell extracts. Thus,

the expression of a carboxyl terminus truncated from of DV2 E promotes its routing to

cell secretion pathways.

Immunogenicity of MV-Dengue 2 recombinant viruses

To rank the immunogenicity of MVvac2-DV2(prME)N and MVvac2-

DV2(prMEsol)N, we immunized huCD46Ge-IFNarKO [123] mice with one or two 105

Page 71: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

60

TCID50 intraperitoneal doses separated by four weeks. As shown in figure 3.3A, measles

neutralization titers of around 1:420 were obtained after one dose of MVvac2 or the two

DV2 E expressing vectors. MV- specific immunity was importantly boosted for MVvac2-

DV2(prMEsol)N which reached neutralizing titers of 1:1,500 14d after the booster dose,

while MVvac2-DV2(prME)N induced only 1:1000 in average. We assessed DV2

neutralization using two related approaches. First, a Log10 neutralization index (LNI),

where the neutralizing effect of individual 1:2 serum dilutions upon a high titer viral DV2

prep infectivity was calculated using averaged preimmune and MVvac2 immunized

serum to determine basal, non-neutralizing conditions. Thus preimmune, MVvac2-

immunized or non-reactive samples tend to have an LNI of zero. Additionally we

determined the classic PRNT50 titer per experimental group using pooled serum samples.

As shown in figure 3.3B, regardless of the immunization schedule, MVvac2-

DV2(prME)N only elicited DV2 neutralization titers just above background. In a

remarkably contrast, MVvac2-DV2(prMEsol)N serum samples reduced the infectivity of

a 105.48 PFU/ml DV2 viral prep by 2.78 and 3.76 orders of magnitude after one or two

doses (equivalent to PRNT50 of 1:160 and 1:214), respectively. Thus, the expression of a

soluble DV2 E glycoprotein and a two-dose schedule outperformed the full-length E

insertion and, expectedly, a single dose immunization scheme.

Generation, characterization and immunogenicity of MVvac2-D4(prMEsol)N

expressing a soluble version of dengue 4 E antigen

Since dengue immunization must elicit a broad neutralizing immunity encompassing

all DV variability, we build upon our DV2-MV findings to generate and characterize an

Page 72: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

61

MV expressing equivalent antigens from DV4. Figure 3.4A depicts the construction of

an MV vectored vaccine expressing a carboxyl-terminus truncated DV4 E glycoprotein.

The recombinant virus, termed MVvac2-DV4(prMEsol)N, replication was assayed by

multistep growth analysis and found to be more similar to MVvac2 than its DV2 Esol

expressing counterpart. DV4 Esol expressing MV reached maximum intracellular titers

of 106.5 TCID50/ml at 48 h post-infection (Fig 3.4B). Similarly, the expression of DV4 E

was corroborated by immunoblot (Fig 3.4C) using specific antibodies against DV4 E and

the expression of MV N as control. The concentration of polyacrylamide (10%) used for

the blot shown did not allow observing a clearer shift in electrophoretic migration of DV4

Esol in comparison with the full-length DV4 E reference. As shown in figure 4E, we

documented DV4 immunogenicity elicited by MVvac2-DV4(prMEsol)N by immunizing

MV-susceptible mice with a two dose IP scheme, as before. As expected, measles

neutralizing immunity was very robust 14 days after the booster dose, reaching titers of

around 1:1,500 (Fig. 3.4E), whereas DV4 neutralizing immunity was also very robust.

Serum samples from immunized animals neutralized (reduced) the infectivity of a high

titer DV4 viral prep (106 PFU/ml) by 4.63 orders of magnitude (DV4 LNI) in average.

This DV4neutralization is equivalent to a 1:640 PRNT50 titer as obtained from pooled

samples. Thus, we were capable of reproducing our MV-DV2 observations using a

similar DV4 vectored antigens.

Page 73: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

62

Immunogenicity of recombinant MV vectors expressing Esol antigens from Dengue

2 and 4

Subsequently, we decided to explore DV2 and DV4 immunogenicity after the

simultaneous administration of MV-DV2 and MV-DV4 Esol vectored vaccines. To this

effect, we immunized MV-susceptible mice groups with either two 105 TCID50 IP doses

of each component for a total measles vaccine dosage of 105.3 per dose (a vaccine scheme

termed “full”) or a 50% reduced dose of each component, 104.7 TCID50 per dose (a

vaccine scheme termed “half”) in order to directly compare the obtained results with our

previous single-component immunizations. Figure 3.5A documents DV2 neutralizing

immunity in mice 14d after two doses of the MV-DV2/4 vectored cocktail. Individual

serum samples had in average a neutralization index of 3.89 and 3.6 Log10 upon a 105.08

PFU/ml DV2 preparation infectivity (equivalent to PRNT50 of 1:430 and 1:240 PRNT50

in pooled serum samples) after full or half dose vaccination schemes, respectively. In a

similar result than the single-component experiment, DV4 neutralizing immunity was

quantitatively higher than DV2 by at least an order of magnitude (Fig 3.5B). The

experimental group had in average an LNI of 5 and 4.38 Log10 upon the DV4 infectivity

of a 106 PFU/ml viral prep (equivalent to PRNT50 of 1:1066 and 1:640 PRNT50 in pooled

serum samples) after full or half dose vaccination schemes, respectively. In average, the

documented measles neutralization immunity was slightly higher than the obtained after

two single-component doses, measles neutralizing titers averaging 1:2,169 and 1:2072,

for full and half vaccination dosage regimes, respectively were documented (data not

shown).

Page 74: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

63

Cross-neutralizing immune responses in MV-susceptible mice inoculated with

MVvac2-D2(prMEsol)N and MVvac2-D4(prMEsol)N

Finally, we explored the neutralizing immunity breadth of our approach by performing

neutralization assays against DV1 and the more phylogenetically distant YFV 17D.

Figure 3.6 documents that serum samples from all of the experimental animals

measurably neutralize these two flaviviruses. The infectivity of a high titer (104.84

PFU/ml) DV1 viral prep was neutralized between 10 to 300 times for an averaged LNI of

2.03 after a full dose vaccination scheme. An almost three times lower titer was obtained

after a half dose vaccination scheme in average. The documented DV1 PRNT50

neutralization titer for full and half dose experimental groups was 1:130 and 1:90,

respectively (Fig 3.6A). Of note, the obtained DV1 immunogenicity after the DV2/4

vaccination cocktail was contrastingly higher than the one obtained after a similar, DV2

single component vaccination (LNI 0.74, PRNT50 1:15) or DV4 single component

vaccination (LNI 1.14, PRNT50 1:36). In a comparable result all of the animals

neutralized YFV. An YFV 17D high titer viral prep (106.78 PFU/ml) infectivity was

neutralized by a factor of 15 to 10 (LNI average of 1.1625 and 0.98) equivalent to a

PRNT50 of 1:60 and 1:40 after full or half vaccination schemes, respectively. Of note, an

LNI of 0.7 is the reported correlate of protection for YFV (equivalent approximately to a

PRNT50 of 1:10) [96]. Again, the obtained YFV 17D immunogenicity after the DV2/4

vaccination cocktail was higher than the one obtained after a similar, DV2 single

component vaccination (LNI 0.056, PRNT50 below detection level) or DV4 single

component vaccination (LNI 0.83, PRNT50 1:12). Thus, by combining DV2 and DV4

Page 75: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

64

insertions we were able to enhance the homologous and heterologous neutralizing

immunity. This effect is not due to transient cross reactivity since single component

immunization did not reach the observed neutralizing titers.

Page 76: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

65

DISCUSSION

In this report we have shown that a fully equivalent, pediatric measles vaccine strain

engineered to express relevant DV2 and DV4 antigens can elicit neutralizing immune

responses in protective ranges against the homologous vectored viral components by the

vaccine cocktail but also against the heterologous DV1 and the relatively distant YFV

17D attenuated strain. We selected the more optimal DV2 antigen to include in our

cocktail. Other studies have suggested the presence of a strong endoplasmic reticulum

retention signal on the transmembrane region of DV2 E protein [127]. This finding

coincides with our observation that a truncated (soluble) DV2 E is significantly more

immunogenic than its full-length counterpart. We have reproduced this observation for

DV4 E immunogenicity also (data not shown). We have tested in-vitro the stability of our

recombinants through passage five, however their genetic stability needs to be assessed

after an in vivo passage. Previous recombinants using the same platform have shown a

robust stability [128].

The enhanced DV neutralizing immunity resulting from the simultaneous

administration of only two serotypes highly suggests that a cocktail formed by the four

DV serotypes will elicit an even more potent DV neutralization. Our selection contains

the representative and significant DV2 and a more distant DV4. The latter is the more

ancient DV serotype, with an E protein sequence closer to other flaviviruses such as JEV

and YFV [129]. A study done by Fu et al., reported that the homology of E-proteins of

DV -1 to -4 share approximately 65%-75% homology at the amino acid level [130, 131].

It remains to be tested experimentally whether the observed broad response is long

Page 77: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

66

lasting, however studies performed in non-human primates with MV vaccine derivatives

point to a long-lived immunity against the vectored antigen [113, 120, 132]. It is

significant, that a robust and simultaneous neutralizing response against two DV

serotypes is usually not associated with enhancement of DV entry and severe forms of

infection. Indeed, hemorrhagic forms associated with a tertiary DV infection are an

extremely rare event [90].

From the practical point of view, the usage of current MV vaccination practices to

deliver DV or flaviviral antigens offer several advantages over other approaches seeking

to supplement the efficiency of current dengue vaccination platforms. First, the individual

pediatric measles dosage is stipulated to contain at least 103 and not more than 104

TCID50 per administered dose[133]. Second, measles protective responses observed in

primates vaccinated with recombinant MV demonstrated vaccine potency equivalence

with the Moraten vaccine strain. Thus, arguably it would be feasible to compose a four

DV serotypes MV vaccine cocktail containing the WHO mandated measles vaccine

dosage to protect one-year-old infants against measles. It would be interesting to explore

the possibility of supplementing the observed MV-DV2 and -DV4 immunogenicity with

prME antigens from other representative mosquito-borne flaviviruses, such as those from

YFV 17D, or Japanese encephalitis virus SA 14-14-2. And thirdly, up-scale production

methods and distribution networks are in place. Vero/hSLAM cells have been the gold

standard to amplify recombinant MV vaccines in the laboratory, however commercial

production requires of the robust fermentation using CEF cells. It is significant then that

we did not observe any effect upon MV-DV2 replication in those cells. Also positive are

Page 78: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

67

the results from the first phase I clinical trial of a MV-Chinkungunya vaccine

documenting the safety and immunogenicity of measles vaccine derivatives [134, 135].

Indeed, Themis Bioscience GmbH has declared intentions for preclinical development of

a MV-Dengue vaccine [136].

There are aspects of our MV-DV approach that need to be experimentally explored.

Importantly, although YFV 17D and measles vaccination does not interfere in African

children that need both vaccinations at the same time[137], DV immune interference

needs to be determined experimentally.

As mentioned, the current dengue vaccination option (DengvaxiaTM, Sanofi Pateur) is

based on chimeric YFV17D virus replication machineries wrapped with the relevant

dengue virus envelope glycoproteins. DV immune responses and protection efficacy after

vaccination is majorly boosted when the recipient’s immune system is primed against

flaviviruses due to a prior exposure to DV, or potentially vaccination against YFV or JEV

in South America or Asia, respectively. Thus, Dengvaxia has been recommended for use

in older than 9 year-old recipients in countries where DV endemicity is close to 80%.

Flavivirus naïve and younger recipients (two to nine-year-old) failed to mount protective

responses against the four dengue serotypes and retrospectively they had a significant

increased risk of hospitalization because of severe dengue infection, [110, 138] . In

highly endemic countries, this vaccination is expected to be safe regardless of the

possibility of having been exposed or not. Indeed, the need to protect young infants from

dengue is very important. The disease is more severe at this age and the intense virus

replication can be the reservoir to sustain an epidemic outbreak [139]. Thus, it would be

Page 79: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

68

very practical to take advantage of the pediatric measles immunization to artificially

prime globally the population against DV or other representative flaviviruses. MV-DV

vaccination would take the place of the primal infection accelerating the start for

immunization with other options and closing the time-gap that would represent the higher

risk. When the time or indication to administer DV, YFV or JEV vaccination presents,

the resulting immune response would be greatly enhanced. Another highly advanced

candidate, spearheaded by Takeda pharmaceuticals, consist of chimeric viruses where the

replication machinery is based on an attenuated DV2. Phase II clinical trial of this

candidate also demonstrated an unbalanced neutralizing immunity against the four DV

serotypes, and most probably will have the same indications upon approval[140]. Thus,

we believe that priming flaviviral immunity at an early age may prove convenient and

complementary to many other flavivirus vaccine approaches.

Page 80: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

69

Page 81: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

70

Figure 3.1. Generation and characterization of recombinant MVs expressing Dengue 2

envelope antigens. (A) Maps of recombinants MVs derived from MVvac2, the parental

strain; MVvac2-D2(prME)N, vectoring prM and a full-length Dengue 2 E glycoprotein

(top) and MVvac2-D2(prMEsol)N expressing a soluble version of the same antigen

(bottom). Both recombinant vectors were obtained from their respective full-length

infectious plasmids by reverse genetics. The native MV coding cistrons are indicated in

gray and the added Dengue 2 antigens in colored boxes. Relevant Dengue 2 E amino

acids and their numbers are indicated. (B) Time-course of cell-associated (top) and cell-

free (bottom) virus production at the indicated time points in Vero/hSLAM cells infected

at an MOI of 0.03 with either the parental strain MVvac2 (continuous line, circles)

MVvac2-D2(prME)N (interrupted line, squares) and MVvac2-D2(prMEsol)N

(interrupted line triangles). Averages and standard deviations of three independent

experiments are shown. (C) Time-course of cell-associated (top) and cell-free (bottom)

virus production at the indicated time points in primary Chick embryo fibroblasts. CEF

cells infected at an MOI of 0.03 with either the parental strain MVvac2 (black bars)

MVvac2-D2(prME)N (grey bars) and MVvac2-D2(prMEsol)N (light grey bars). Average

of a representative experiment performed in triplicate are shown. (D) Viral plaques

morphology in Vero/hSLAM cells obtained 96h post-infection and stained with naphtol

blue-black.

Page 82: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

71

Page 83: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

72

Figure 3.2. Expression of Dengue 2 E glycoprotein by recombinant viruses. (A).

Vero/hSLAM cells infected at an MOI of 0.5 were mock- infected or either with the

parental strain MVvac2 , MVvac2-D2(prME)N and MVvac2-D2(prMEsol)N. At 48h

post-infection, infected cells were fixed and stained with fluorescent labeled antibodies

and nuclei counterstained with DAPI (blue channel). Single channel and overlaid images

after epifluorescence microscopy are shown, red channel (anti-MV reactivity) and green

channel (anti dengue 2 prM and E), white bar represents 200nm. (B) Analysis of

expression by Western blot of proteins extracted from MVvac2- , MVvac2-D2(prME)N-

and MVvac2-D2(prMEsol)N-, or mock-infected cells. Proteins were detected using

antibodies against dengue 2 E, MV N, and actin, and the positions of molecular weight

standards are indicated to the left. Left panel, analysis of the glycosylation of the vectored

Dengue 2 E glycoprotein. Lysates from cells infected with MVvac2, MVvac2-

D2(prME)N and MVvac2-D2(prMEsol)N were treated or not with the indicated

glycosidase and analyzed by western blot using anti MV-H (top panel) and anti Dengue 2

E (bottom panel) antibodies. Endo-H resistant forms are indicated with asterisks.

Molecular weight standard positions are indicated to the left.

Page 84: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

73

Page 85: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

74

Figure 3.3. Immunogenicity of MV-Dengue 2 recombinant viruses. MV-susceptible mice

received one or two IP doses of MVvac2 , MVvac2-D2(prME)N and MVvac2-

D2(prMEsol)N. Neutralizing immune responses were analyzed six weeks after the first

dose. (A) Anti MV neutralization assessed by PRNT90. (B) Anti-Dengue neutralization

assessed by Log10 Neutralization Index (LNI, left panel) or PRNT50 performed in

triplicate using pooled serum samples. Each symbol represents one animal; horizontal

lines and vertical brackets indicate group average and standard deviation, respectively.

Anti-Dengue 2 PRNT50 titers are indicated.

Page 86: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

75

Page 87: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

76

Figure 3.4. Generation, characterization and immunogenicity of MVvac2-D4(prMEsol)N

expressing a soluble version of dengue 4 E antigen. (A) Map of MVvac2-D4(prMEsol)N.

Native MV coding cistrons are indicated in gray and the added Dengue 4 prM and Esol

antigens in colored boxes. Relevant Dengue 4 E amino acids and their numbers are

indicated. (B) Time-course of cell-associated virus production at the indicated time points

in Vero/hSLAM cells infected at an MOI of 0.03 with either the parental strain MVvac2

(continuous line, circles) and MVvac2-D4(prMEsol)N (interrupted line, diamonds).

Averages and standard deviations of three independent experiments are shown. (C)

Expression of Dengue E protein demonstrated by Western blot. Cell lysates of infected

cells (inoculum indicated on top) were probed with anti MV N, anti Dengue E or anti-

actin antibodies. The relative migration of molecular weight markers is indicated on left.

(D). MV-susceptible mice received two IP doses of MVvac2 and MVvac2-

D4(prMEsol)N. Neutralizing immune responses were analyzed two weeks after the

booster dose. (A) Anti MV neutralization assessed by PRNT90. (B) Anti-Dengue

neutralization assessed by Log10 Neutralization Index (LNI, left panel) or PRNT50

performed in triplicate using pooled serum samples (right panel). Each symbol represents

one animal; horizontal lines and vertical brackets indicate group average and standard

deviation, respectively. Anti-Dengue 4 PRNT50 titers are indicated.

Page 88: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

77

Page 89: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

78

Figure 3.5. Immunogenicity of recombinant MV vectors expressing Esol antigens from

Dengue 2 and 4. MV-susceptible mice received two IP doses of MVvac2-D2(prMEsol)N

and MVvac2-D4(prMEsol)N in a 28 day interval. Neutralizing immune responses were

analyzed two weeks after the booster dose. (A) Analysis of neutralization against Dengue

2 and (B) against Dengue 4 viruses. Anti dengue neutralizing immune responses were

assessed by LNI (left panel) and PRNT50 (right panel). Each symbol represents one

animal; horizontal lines and vertical brackets indicate group average and standard

deviation, respectively. Averages of anti-Dengue PRNT50 titers determined in triplicate of

pooled serum samples are indicated.

Page 90: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

79

Page 91: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

80

Figure 3.6. Cross-neutralizing immune responses in MV-susceptible mice inoculated

with MVvac2-D2(prMEsol)N and MVvac2-D4(prMEsol)N (A) Analysis of

neutralization against Dengue 1 and (B) against attenuated YFV. Anti dengue and anti-

YFV neutralizing immune responses were assessed by LNI (left panel) and PRNT50 (right

panel). Each symbol represents one animal; horizontal lines and vertical brackets indicate

group average and standard deviation, respectively. Averages of anti-Dengue PRNT50

titers determined in triplicate of pooled serum samples are indicated.

Page 92: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

81

REFERENCES

1. Guzman, M.G. and E. Harris, Dengue. The Lancet, 2015. 385(9966): p. 453-465. 2. Martina, B.E., P. Koraka, and A.D. Osterhaus, Dengue virus pathogenesis: an

integrated view. Clinical microbiology reviews, 2009. 22(4): p. 564-81. 3. Lisova, O., et al., Mapping to completeness and transplantation of a group-

specific, discontinuous, neutralizing epitope in the envelope protein of dengue virus. Journal of General Virology, 2007. 88(9): p. 2387-2397.

4. Schieffelin, J.S., et al., Neutralizing and non-neutralizing monoclonal antibodies

against dengue virus E protein derived from a naturally infected patient. Virology journal, 2010. 7: p. 28.

5. Wahala, W.M., et al., Dengue virus neutralization by human immune sera: role of

envelope protein domain III-reactive antibody. Virology, 2009. 392(1): p. 103-13. 6. Henchal, E.A. and J.R. Putnak, The dengue viruses. Clinical microbiology

reviews, 1990. 3(4): p. 376-96. 7. Schwartz, L.M., et al., The dengue vaccine pipeline: Implications for the future of

dengue control. Vaccine, 2015. 33(29): p. 3293-3298. 8. Beltramello, M., et al., The human immune response to Dengue virus is

dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity. Cell host & microbe, 2010. 8(3): p. 271-83.

9. Capeding, M.R., et al., Clinical efficacy and safety of a novel tetravalent dengue

vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. The Lancet, 2014. 384(9951): p. 1358-1365.

10. Institute, B. Phase III Trial to Evaluate Efficacy and Safety of a Tetravalent

Dengue Vaccine. July 21, 2016 [cited 2016 08/08/2016]; Available from: https://clinicaltrials.gov/ct2/show/NCT02406729.

11. Guy, B., et al., Development of the Sanofi Pasteur tetravalent dengue vaccine:

One more step forward. Vaccine, 2015. 33(50): p. 7100-7111. 12. Halstead, S.B. and P.K. Russell, Protective and immunological behavior of

chimeric yellow fever dengue vaccine. Vaccine, 2016. 34(14): p. 1643-1647. 13. Villar, L., et al., Efficacy of a tetravalent dengue vaccine in children in Latin

America. New England Journal of Medicine, 2015. 372(2): p. 113-123.

Page 93: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

82

14. Perry, R.T., et al., Progress toward regional measles elimination-worldwide,

2000-2013. MMWR Morb Mortal Wkly Rep, 2014. 63(45): p. 1034-8. 15. Brandler, S., et al., Pediatric measles vaccine expressing a dengue antigen

induces durable serotype-specific neutralizing antibodies to dengue virus. PLoS neglected tropical diseases, 2007. 1(3): p. e96.

16. Brandler, S., et al., Pediatric measles vaccine expressing a dengue tetravalent

antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine, 2010. 28(41): p. 6730-9.

17. Brandler, S. and F. Tangy, Recombinant vector derived from live attenuated

measles virus: potential for flavivirus vaccines. Comparative immunology, microbiology and infectious diseases, 2008. 31(2-3): p. 271-91.

18. Del Valle, J.R., et al., A vectored measles virus induces hepatitis B surface

antigen antibodies while protecting macaques against measles virus challenge. Journal of virology, 2007. 81(19): p. 10597-10605.

19. Ono, N., et al., Measles viruses on throat swabs from measles patients use

signaling lymphocytic activation molecule (CDw150) but not CD46 as a cellular receptor. Journal of virology, 2001. 75(9): p. 4399-4401.

20. Tatsuo, H., N. Ono, and Y. Yanagi, Morbilliviruses use signaling lymphocyte

activation molecules (CD150) as cellular receptors. Journal of virology, 2001. 75(13): p. 5842-5850.

21. Radecke, F. and M. Billeter, Appendix: measles virus antigenome and protein

consensus sequences, in Measles Virus1995, Springer. p. 181-192. 22. Bredenbeek, P.J., et al., A stable full-length yellow fever virus cDNA clone and

the role of conserved RNA elements in flavivirus replication. Journal of general virology, 2003. 84(5): p. 1261-1268.

23. Radecke, F., et al., Rescue of measles viruses from cloned DNA. The EMBO

journal, 1995. 14(23): p. 5773. 24. Parks, C.L., et al., Enhanced measles virus cDNA rescue and gene expression

after heat shock. Journal of virology, 1999. 73(5): p. 3560-3566. 25. Karber, G., 50% end-point calculation. Arch Exp Pathol Pharmak, 1931. 162: p.

480-483.

Page 94: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

83

26. Reyes-del Valle, J., et al., Protective anti-hepatitis B virus responses in rhesus monkeys primed with a vectored measles virus and boosted with a single dose of hepatitis B surface antigen. Journal of virology, 2009. 83(17): p. 9013-9017.

27. Bhamarapravati, N., et al., Immunization with a live attenuated dengue-2-virus

candidate vaccine (16681-PDK 53): clinical, immunological and biological responses in adult volunteers. Bulletin of the World Health Organization, 1987. 65(2): p. 189.

28. Halstead, S.B. and P. Simasthien, Observations related to the pathogenesis of

dengue hemorrhagic fever. II. Antigenic and biologic properties of dengue viruses and their association with disease response in the host. The Yale journal of biology and medicine, 1970. 42(5): p. 276.

29. Mrkic, B., et al., Measles virus spread and pathogenesis in genetically modified

mice. Journal of virology, 1998. 72(9): p. 7420-7427. 30. de Swart, R.L., S. Yüksel, and A.D. Osterhaus, Relative contributions of measles

virus hemagglutinin-and fusion protein-specific serum antibodies to virus neutralization. Journal of virology, 2005. 79(17): p. 11547-11551.

31. Zuniga, A., et al., Attenuated measles virus as a vaccine vector. Vaccine, 2007.

25(16): p. 2974-2983. 32. Tangy, F. and H.Y. Naim, Live attenuated measles vaccine as a potential

multivalent pediatric vaccination vector. Viral immunology, 2005. 18(2): p. 317-326.

33. Mason, R.A., et al., Yellow fever vaccine: direct challenge of monkeys given

graded doses of 17D vaccine. Applied microbiology, 1973. 25(4): p. 539-544. 34. Hsieh, S.-C., et al., A strong endoplasmic reticulum retention signal in the stem–

anchor region of envelope glycoprotein of dengue virus type 2 affects the production of virus-like particles. Virology, 2008. 374(2): p. 338-350.

35. Devaux, P., et al., Attenuation of V-or C-defective measles viruses: infection

control by the inflammatory and interferon responses of rhesus monkeys. Journal of virology, 2008. 82(11): p. 5359-5367.

36. Lanciotti, R.S., D.J. Gubler, and D.W. Trent, Molecular evolution and phylogeny

of dengue-4 viruses. Journal of General Virology, 1997. 78(9): p. 2279-2284.

Page 95: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

84

37. Venkatachalam, R. and V. Subramaniyan, Homology and conservation of amino acids in E-protein sequences of dengue serotypes. Asian Pacific Journal of Tropical Disease, 2014. 4: p. S573-S577.

38. Fu, J., et al., Full-length cDNA sequence of dengue type 1 virus (Singapore strain

S275/90). Virology, 1992. 188(2): p. 953-958. 39. Combredet, C., et al., A molecularly cloned Schwarz strain of measles virus

vaccine induces strong immune responses in macaques and transgenic mice. Journal of virology, 2003. 77(21): p. 11546-11554.

40. Kosasih, H., et al., Report of four volunteers with primary, secondary and tertiary

dengue infections during a prospective cohort study. Dengue Bulletin, 2006. 30: p. 87.

41. Anonymous, Measles Potency test. Code of Federal regulation, CFR Title 21.Sec

630.34., April 1, 1996. 42. Ramsauer, K., et al., Immunogenicity, safety, and tolerability of a recombinant

measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. The Lancet infectious diseases, 2015. 15(5): p. 519-527.

43. Brandler, S., et al., A recombinant measles vaccine expressing chikungunya virus-

like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine, 2013. 31(36): p. 3718-3725.

44. THEMIS. THEMIS. NEW HORIZONS FOR VACCINES. 2016 [cited 2016

9/2/2016]; Available from: http://www.themisbio.com/ - /products-and-pipeline. 45. Stefano, I., et al., Recent immunization against measles does not interfere with the

sero-response to yellow fever vaccine. Vaccine, 1999. 17(9): p. 1042-1046. 46. Ferguson, N.M., et al., Benefits and risks of the Sanofi-Pasteur dengue vaccine:

Modeling optimal deployment. Science, 2016. 353(6303): p. 1033-1036. 47. Guzmán, M.G., et al., Effect of age on outcome of secondary dengue 2 infections.

International journal of infectious diseases, 2002. 6(2): p. 118-124. 48. Takeda. Safety and Immunogenicity With Two Different Serotype 2 Potencies of

Tetravalent Dengue Vaccine (TDV) in Adults in Singapore. 2016 [cited 2016 08/08/2016]; Available from: https://clinicaltrials.gov/ct2/show/study/NCT02425098?term=dengue+and+takeda&rank=4.

Page 96: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

85

CHAPTER 4 GENERATION OF A RECOMBINANT MEASLES VIRUS EXPRESSING FULL-

LENGTH DENGUE 4 ENVELOPE GLYCOPROTEINS

ABSTRACT

Dengue virus (DV), an arthropod-borne viral disease, causes major public health

concern in the world due to a large number of people infected and the economic impact

for endemic countries. There have been multiple attempts to generate a vaccine to prevent

infection by any of the four described DV serotypes. A recently approved vaccine

demonstrated disease enhancement in young children associated with a flavivirus naïve

status. Therefore, a pediatric vaccination strategy with the potential to prime neutralizing

anti-flaviviral responses in one-year-old infants can be used to complement vaccine

immunity to protect young children. In a previous work, we modified a measles vaccine

viral vector (MVvac2) to express the pre membrane (prM) and a full length or a truncated

(90%) form of the major envelope (E) from DV2, and we showed that a truncated DV2 E

isoform has superior immunogenicity to its coserponding full-length construct. Here, we

generated a recombinant measles virus expressing DV4 PrM and full-length E protein.

The recombinant MV-D4-PrME vaccine elicited robust anti-measles neutralizing

responses (1: 1280 TCID50/ml in average) and a very discreye homologous anti-dengue

4 neutralizing responses in all MV-susceptible mice 1:18 PRNT50 in average). In sum,

we were able to reproduce the poor immunogenicity of a DV2 envelope glycoprotein

antigen when expressed at full-length in the more distant DV4 background. It is possible

that soluble versions of flaviviral envelope antigens are more immunogenic despite the

Page 97: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

86

presence or not of endoplasmic reticulum retention signals in its transmembrane region,

as it is the case for DV2.

Page 98: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

87

INTRODUCTION

Dengue virus envelope glycoprotein is the major antigen in the surface of the

virion and a biomarker of evolution of the Dengue serogroup. The phylogenetic

difference in E sequence between DV 1 to DV 4 is 25-35%, mainly due to differences in

domain III of its ectodomain. The transmembrane (TM) region of DV glycoproteins E

and PrM is of paramount importance to consider because it may contain endoplasmic

reticulum ER sequestering regions. The presence of the ER localization signals in the

flavivirus envelope proteins is essential for the virion morphogenesis of the flaviviruses.

That occurs in association with ER membranes, suggesting that there should be

accumulation of the virion components in this compartment[129] [141]. Flavivirus

acquire their envelope by budding through ER membranes, therefore the E protein needs

to accumulate in the ER compartment before budding can take place. The secretion of

truncated E protein of flavivirus with its C-terminal transmembrane removed suggested

that there is no ER retention signal in the ectodomain of this protein. Some studies

showed that transmembrane domanine of E protein from YFV and DV2 contain strong

retention signal. When the ectodomain of CD4 fused with the TM domain of E from DV2

or YFV, it was retained in the ER. Indicating that the TM domains of flavivirus envelope

proteins contains ER retention signal.

The envelope E protein is essential for vaccine development as it has many

epitopes that react with neutralizing antibodies. Envelope protein is a glycoprotein, which

has been studied for its antigenicity and immunogenicity[142, 143]. The presence of this

retention signal in the transmembrane region may result in poor immunignicity of viral

Page 99: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

88

vectors expressing the full length E protein. In the pervious work we showed that

recombinant measles expressing full length E protein generate poor anti-DV2 neutralzing

antibodies. This finding suggested that presence of ER retention signal contributed to the

poor immunity due to inefficient antigen presentation.

The correct folding of E protein is critical determine of immunogenicity. The co-

expression of Pre membrane PrM protein is vital for proper folding as it acts as a scaffold

for the E protein[144]. We constructed a vaccine candidate based on measles vaccine

virus expressing pre-membrane (prM) and envelope (E) proteins of dengue 4 virus (DV-

4) and tested it in mice to evaluate immunogenicity and protection against DV-4

infection. Measles has been successfully used as vaccine vectors for several viruses to

induce strong humoral and cellular immune responses.

Page 100: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

89

MATERIAL AND METHODS

Cells and Viruses.

Vero/hSLAM cells [114, 115]were maintained in Dulbecco’s modified Eagle’s

medium with high glucose concentration (DMEM, Sigma-Aldrich, St. Louis, MO, USA)

supplemented with 5% fetal bovine serum (FBS, Atlanta Biologicals, Flowery Branch,

GA, USA), 1% penicillin–streptomycin (Sigma-Aldrich) and 0.5 mg/mL G418 (Enzo

Life Sciences, Farmingdale, NY, USA) to maintain hSLAM expression selection. BHK

21 cells were maintained in DMEM supplemented with 10% FBS and 1% penicillin-

streptomycin. Helper 293-3-46 cells [116]a derivative from HEK 293 cells were

maintained in DMEM with 10% FBS, 1% PS, and 1.2 mg/mL G418.

DV4 virus was obtained from the ATCC (VR-1257 strain H241 and propagated in

Vero cells. The virus was then amplified in BHK21 cells for up to two or three passages.

Recombinant MVs were generated using the method of Radecke et al. [145]

modified by Parks et al. [118] After detection of their cytopathic effect on mixed cultures

of Helper 293-3-46 and Vero/hSLAM cells, single syncytia were transferred to and

propagated in Vero/hSLAM cells. Viral clones’ stocks were amplified by infecting

Vero/hSLAM cells at a multiplicity of infection (MOI) of 0.03 and incubated at 37°C.

When approximately 80% cytopathic effect was observed, cells were scraped in Opti-

MEM (Life Technologies, Grand Island, NY) and viral particles were released by two

freeze-thaw cycles.

Multi-step growth kinetics of the viruses were determined by infecting

Vero/hSLAM cells at an MOI of 0.03 and incubating them at 37°C. Infected cells and

Page 101: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

90

supernatants were collected and lysed by a single cycle of freeze-thaw at prescribed times

post-infection, and the 50% tissue culture infectious dose (TCID50) was determined in

Vero/hSLAM using the Spearman-Kärber end-point dilution method. Primary chick

embryo fibroblast (CEF) were obtained from ATCC (CRL-1590 F2) and grown in

Dulbecco’s modified Eagle’s medium with high glucose concentration (DMEM, Sigma-

Aldrich, St. Louis, MO, USA) supplemented with 10% FBS, 1% penicillin–streptomycin

and 10% tryptose phosphate broth (Gibco, Life Technologies). Kinetics of infection of

CEF were performed by infecting CEF at MOI of 0.03 with MV and DV-expressing

recombinant MV and collecting cell-associated and supernatant at the indicated time

post-infection.

Plasmids and plasmids constructions

The plasmid pB(+)MVvac2(HBsAg)N was used to construct the recombinant

MVs expressing DV4 antigens[120]. This plasmid contains an infectious MV cDNA

corresponding to the antigenome of the Schwarz measles vaccine strain with an

additional transcription unit (ATU) inserted downstream of the nucleocapsid (N) cistron

to direct the expression of an inserted foreign gene. To generate the recombinant measles

expressing DV4 prM- E glycoproteins, MluI and AatII digestion fragments containing the

desired coding sequence were obtained by PCR (primer sequence available upon request)

and swapped with the HBsAg insert. DV4 prME coding sequences have been achieved

from the full-length pR424-D4 plasmid (H241 strain). The full-length recombinant

plasmid was termed pB(+)MVvac2-DV4(prME)N. Correct insertions and relevant gene

boundaries were corroborated by sequencing.

Page 102: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

91

Indirect Immunofluorescence Microscopy

Vero/hSLAM cells were seeded into 4 well glass Millicell EZ slides

(MILLPORE) and infected at MOI of 0.06, 0.1 and 1 with MV, MVvac2DV4(PrME)N,

and DV4, respectively. After 48 hours of incubation at 37 C, the cells were fixed with 3%

paraformaldehyde and incubated at 37 °C overnight. The cells were permeabilized with

0.1% Triton X-100 in phosphate- buffer saline (PBS). Both primary and secondary

antibodies dilution were prepared in 0.1% Triton X-100 in 10% FBS serum in 1 X PBS.

Two Mixes of primary antibodies were used, monoclonal antibody directed against DV

1,2,3,4 E protein at 1:50 dilution (a mouse IgG2a cat. # 76 ab9202 from abcam) or

monoclonal antibody directed against DV1,2,3,4 PrM protein at 1:50 dilution (a mouse

IgG cat.# ab41473 from abcam) with the polyclonal antibodies directed against MV N

protein at 1:300 dilution (a rabbit IgG antibody provided from R. Cattaneo). The primary

antibodies incubations were carried out overnight at 4 °C. A mix of goat anti-mouse IgG

coupled to Fitc (Green fluorescence), and a goat anti-rabbit IgG coupled to PE (Red

fluorescence) were used as a secondary antibody at dilution of 1:300 and incubated at

room temperature for 2 hours. The coverslips were mounted on slides by using

flouromount G with DAPI (Electron Microscopy Science). The slides examined, and

images were taken by an inverted microscope with epifluorescence illumination

(EVOSFL).

Mice inoculations

All experimental procedures were performed according to a protocol approved by

the Arizona State University Institutional Animal Care and Use Committee. To determine

Page 103: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

92

the immunogenicity of MVvac2DV4(PrME)N in an MV susceptible small animal host,

groups of HuCD46Ge-IFNarKO mice were inoculated by the intraperitoneal (i.p.) route

with the indicated doses diluted in 100 uL of Opti-MEM[120]. Two weeks after the

initial inoculation, the mice received a similar booster dose. One week after the second

dose, mice were exsanguinated. Sera were separated and heat-inactivated at 56°C for 1 h.

MV neutralization assay

To determine MV neutralization titer, serial two-fold dilutions of heat-inactivated

sera in Opti-MEM were incubated with 100 TCID50 of MVvac2 for 1 h at 37 °C.

Vero/hSLAM cells were added to the serum–virus mixtures and the plates were then

incubated at 37 °C for three days, at which time neutralization titer was assessed as the

highest dilution of serum capable of complete neutralization of infectivity, defined as the

absence of MV cytopathic effect. Neutralization titers were reported as the average of a

determination made in triplicate.

DV neutralization assay

To determine DV4 Log10 Neutralization Index titer the corresponding viral prep

was mixed with heat-inactivated serum in a 1:1 proportion in a 25 uL volume of Opti-

MEM and incubated for 1 h at 37 °C. Infectivity after serum incubation was determined

using a plaque assay in BHK-21 cells. Briefly, ten-fold serial dilutions of the virus/serum

mixtures were generated using Opti-MEM and added to 2.4 × 106 BHK-21 cells in a 24-

well plate and then incubated for 4 h at 37 °C. At this time, 500 uL of 3% carboxymethyl

cellulose was added to each well and plates were incubated for 5 days at 37 °C. The wells

were then washed; the monolayer was fixed and stained with acetic acid/naphtol blue-

Page 104: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

93

black and plaques were counted. The neutralization index was expressed as the

logarithmic difference between the averaged DV4 titer when incubated with control,

namely, that of animals inoculated with MVvac2, and preimmune. Neutralization indexes

thus determined were graphed as Log10NI.

In another approach, we performed a standard 50% plaque reduction

neutralization assay (PRNT50) using sera from each animal. Briefly, 50 DV4 PFU were

incubated with two-fold serum dilutions (from 1:5 to 1:320) for 1 h at 37 °C. Remaining

infectivity was determined by plaque assay, as described above. The neutralization titer

was defined as the dilution that resulted in a 50% reduction in plaque numbers, as

compared to unrelated serum.

RESULTS

Generation of recombinant divalent MV-DV4 vaccine vector.

We modified a full-length infectious cDNA plasmid named pB(+)MVvac2. This

plasmid and its derivatives can be used to recover recombinant MVs by well-established

reverse genetic techniques. DV4 prME coding sequences were inserted downstream MV

N gene. The coding sequence consisted of prM and E full-length coding sequences

preceded by the signal sequence of the carboxyl terminus of the capsid protein (C

anchor). The recovered virus was termed MVvac2-DV4(prME)N and expresses intact

full-length prM-E glycoproteins. (Fig4.1A). To corroborate the expression of DV4

vectored antigens, we performed an indirect immunofluorescence assays using antibodies

specific for MV-N and DV4 PrM or E (stained with secondary RFP and GFP conjugated

antibodies, respectively). To track the viral cytopathic effect, cell nuclei were

Page 105: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

94

counterstained with DAPI. As evident in Figure 4.1B, merged epifluorescence images

from cells infected with either MVvac2-DV4(prME)N had a strong yellow

epifluorescence resulting from co-localization of green and red signals coming from

bonafide cell syncitia induced by viral infection. In contrast, control mock infected cells

showed no green or red epifluorescence, respectively. Furthermore, plaque morphology

analysis revealed that MVvac2 and MVvac2-DV4(prME)N had circular plaques of

around 1.082 and 1.079 mm in diameter respectively (Figurer 4.1D).

Replication fitness of recombinant MV-DV4 is equivalent to parental MV.

To corroborate the effect of DV4 prME insertion upon recombinant MV

replication fitness, we performed multistep growth analysis using two cell substrates.

Initially, viral replication in Vero/hSLAM did not demonstrate a major deleterious effect

because of the genetic modification on MV backbone. Intracellular MVvac2-

DV4(prME)N viral yields reached 6.8 Log10 TCID50/ml at 48h PI, which was slightly

higher than but not significantly different of the parental strain MVvac2. The

extracellular fractions had similar patterns of replication with viral yields diminished by

one log10 approximately in comparison to the intracellular fraction (Fig4.2A &B). The

recombinant MV-DV4 is attenuated MV strain (Moraten/Schwarz) derivatives; we

analyzed its replication in a cell line used for commercial production, primary chicken

embryo fibroblasts. In these cells we observed a very similar replication profile of Mv-

DV4when compared to the parental MVvac2, reaching maximum intracellular titers of

around 5 log10 TCID50/ml at 120 hours PI and extracellular titers of 2.5 log10

TCID50/ml (Fig4.2C&D).

Page 106: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

95

Recombinant MV-DV4 induces anti-measles and anti- dengue 4 immune responses.

We immunized huCD46Ge-IFNarKO mice with two 105 TCID50 intraperitoneal

doses separated by three weeks. As shown in figure 4.3A, measles neutralization titers of

around 1:420 were obtained after one dose of MVvac2. MV- specific immunity was

boosted for two doses of MVvac2-DV4(prME)N which reached neutralizing titers of 1:

1280. We assessed DV4 neutralization using two related approaches. First, a Log10

neutralization index (LNI), where the neutralizing effect of individual 1:2 serum dilutions

upon a high titer viral DV4 prep infectivity was calculated using averaged preimmune

and MVvac2 immunized serum to determine basal, non-neutralizing conditions. Thus

preimmune, MVvac2-immunized or non-reactive samples tend to have an LNI of zero.

Additionally, we determined the classic PRNT50 titer per experimental group using

pooled serum samples. As shown in figure 4.3B, MVvac2-DV4(prME)N only elicited

DV4 neutralization titers just above background with average 0.5 LNI and 1: 18

PRNT50.

DISCUSSION

There have been challenges to develop a dengue vaccine. The results of a phase 3

clinical trial of the the Sanofi Pasteur yellow fever/dengue chimeric vaccine “CYD” in

children of the Asia-Pacific and Latin America regions, failed to protect against DV2 and

showed limited protection against DV1, including the enhancement of disease and the

increased risk of hospitalization of children under 9 year old, indicating the need to

pursue new strategies to reach a broader protection against dengue infections[83]. The

recombinant live attenuated viruses are attractive strategies to construct dengue vaccines

Page 107: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

96

because they can induce strong humoral responses that mimic natural infection and are

safe, low coast vaccine candidates[14-16, 22]. However, it is very critical to expresses the

antigen in forms that will generates a robust immune response.

In this study, we describe, the construction of measles vectoring dengue 4-

glycoprotein candidate vaccine. Our recombinant measles expresses prM and E proteins

of DV-4 as was demonstrated by immunofluorescent. The replication fitness of the

recombinant virus was comparable to the parental strain as it was tested in two different

cell types. Our MV-DV4-prME induces anti- MV and anti-DV4 humoral response in

HuCD46Ge-IFNarKO mice. Inducing neutralizing antibodies is the main target of

vaccine candidates as it blocks virus entry into target cells. Therefore, we explored the

construction of dengue vaccines by expressing full-length E protein to induce

neutralizing antibody which can bind these proteins and prevent it from binding to host

cell receptors. However, our data show weak neutralizing immune response against

dengue 4 virus (average 1:18 PRNT50). In our previous work we generated recombinant

measles expressing both prM and truncated DV4 E protein, which produces strong anti-

DV4 neutralizing response, the difference in the immunogenicity between the two

recombinant measles can be explained by different antigen presentations of the full-

length and truncated E protein. This data suggested that soluble versions of flaviviruses E

could be a better candidate for construction of anti-falvivirus vector vaccines. Flavivirus

envelope proteins TM lack any of the classical ER retrieval signals, KDEL, KKXX, or

RRXX , the two helices TM1 and TM2 in the transmembrane region of the four DV

Page 108: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

97

serotypes and YFV share identical amino acids resdiues which may play a role in the

retention of the E proteins in the ER(Figure4.4).

In future work, we will compare the enhancement of DV1 and DV2 infection in

macrophage cell line (P388 D1) between the serum of animal inoculated by the truncated

or full-length DV4 recombinant measles.

Page 109: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

98

Page 110: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

99

Figure4.1. Generation and characterization of recombinant MVs expressing Dengue 4 PrM and E antigens. (A) Maps of recombinants MVs derived from MVvac2, the parental strain; MVvac2-D4(prME)N, vectoring prM and a full-length Dengue 4 E glycoprotein. Recombinant MV is obtained from its respective full-length infectious plasmids by reverse genetics. The native MV coding cistrons are indicated in gray and the added Dengue 4 antigens in colored boxes. Relevant Dengue 4 E amino acids and their numbers are reported. (B) Expression of Dengue 4 PrM or E proteins by recombinants MV vectors. Vero/hSLAM cells infected at an MOI of 0.5 are mock- infected or MVvac2-D4(prME)N infected. At 48h post-infection, infected cells were fixed and stained with fluorescent-labeled antibodies and nuclei counterstained with DAPI (blue channel). Overlaid images after epifluorescence microscopy are shown, red channel (anti-MV reactivity) and green channel (anti-dengue 4 prM or E), white bar represent 200nm. (C) Viral plaques morphology in Vero/hSLAM cells obtained 96h post-infection and stained with naphtol blue-black.

Page 111: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

100

Page 112: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

101

Figure4.2. Replication finesses of recombinant MV-DV4. Time-course of cell-associated (A) and cell-free (B) virus production at the indicated time points in Vero/hSLAM cells infected at an MOI of 0.03 with either the parental strain MVvac2 (continuous line, circles) MVvac2-D4(prME)N (continuous line, squares) Averages and standard deviations of three independent experiments are shown. (C) Time-course of cell-associated and (D) cell-free virus production at the indicated time points in primary Chick embryo fibroblasts. CEF cells infected at an MOI of 0.03 with either the parental strain MVvac2 (dark grey bars) MVvac2-D4(prME)N (grey bars). Averages and standard deviations of three independent experiments are shown.

Page 113: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

102

Figure4.3. Immunogenicity of the MV-DV4 recombinant virus. MV-susceptible mice received one IP dose of MVvac2 or two IP doses MVvac2-D4(prME)N. Neutralizing immune responses were analyzed six weeks or 4 weeks after the first dose. (A) Anti MV neutralization assessed by PRNT90. (B) Anti-Dengue neutralization evaluated by Log10 Neutralization Index (LNI, left panel) or PRNT50 performed in triplicate using pooled serum samples. Each symbol represents one animal; horizontal lines and vertical brackets indicate group average and standard deviation, respectively. Anti-Dengue 4 PRNT50 titers are shown.

Page 114: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

103

Figure 4.4. Sequance alignment of TM regions of E protein from DV1-4 and YFV. DV1(Genbank accession number FJ687432), DV2(Genbank accession number NC_001474), DV3 (Genbank accession number FJ850055), DV4 (Genbank accession number AY618990), YFV (Genbank accession number AY640589). Identical amino acid residues are highlighted in tallow. The predicted TM1 and TM2 helices are indicated at the bottom. Adapted from [1]

Page 115: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

104

REFRENCES

1. Gubler, D.J., Dengue and dengue hemorrhagic fever. Clinical microbiology reviews, 1998. 11(3): p. 480-96.

2. Gubler, D.J., Epidemic dengue/dengue hemorrhagic fever as a public health,

social and economic problem in the 21st century. Trends in microbiology, 2002. 10(2): p. 100-3.

3. Guzman, M.G., et al., Dengue: a continuing global threat. Nature Reviews Microbiology, 2010. 8: p. S7-S16.

4. Schwartz, L.M., et al., The dengue vaccine pipeline: Implications for the future of

dengue control. Vaccine, 2015. 33(29): p. 3293-3298. 5. Lanciotti, R.S., D.J. Gubler, and D.W. Trent, Molecular evolution and phylogeny

of dengue-4 viruses. Journal of General Virology, 1997. 78(9): p. 2279-2284. 6. Wang, E., et al., Evolutionary relationships of endemic/epidemic and sylvatic

dengue viruses. Journal of virology, 2000. 74(7): p. 3227-3234. 7. Gubler, D.J., Dengue and dengue hemorrhagic fever. Clinical microbiology

reviews, 1998. 11(3): p. 480-496. 8. Foster, J.E., et al., Phylogeography and molecular evolution of dengue 2 in the

Caribbean basin, 1981–2000. Virology, 2004. 324(1): p. 48-59. 9. Jenkins, G.M., et al., Evolution of base composition and codon usage bias in the

genus Flavivirus. Journal of molecular evolution, 2001. 52(4): p. 383-390. 10. Kuno, G., et al., Phylogeny of the genus Flavivirus. Journal of virology, 1998.

72(1): p. 73-83. 11. Chen, Y., et al., Dengue virus infectivity depends on envelope protein binding to

target cell heparan sulfate. Nature medicine, 1997. 3(8): p. 866-871. 12. Whitehead, S.S., et al., Prospects for a dengue virus vaccine. Nature Reviews

Microbiology, 2007. 5(7): p. 518-528. 13. Li, L., et al., The flavivirus precursor membrane-envelope protein complex:

structure and maturation. Science, 2008. 319(5871): p. 1830-1834. 14. Ono, N., et al., Measles viruses on throat swabs from measles patients use

signaling lymphocytic activation molecule (CDw150) but not CD46 as a cellular receptor. Journal of virology, 2001. 75(9): p. 4399-4401.

Page 116: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

105

15. Tatsuo, H., N. Ono, and Y. Yanagi, Morbilliviruses use signaling lymphocyte

activation molecules (CD150) as cellular receptors. Journal of virology, 2001. 75(13): p. 5842-5850.

16. Radecke, F. and M. Billeter, Appendix: measles virus antigenome and protein

consensus sequences, in Measles Virus1995, Springer. p. 181-192. 17. Radecke, F., et al., Rescue of measles viruses from cloned DNA. The EMBO

journal, 1995. 14(23): p. 5773. 18. Parks, C.L., et al., Enhanced measles virus cDNA rescue and gene expression

after heat shock. Journal of virology, 1999. 73(5): p. 3560-3566. 19. Reyes-del Valle, J., et al., Protective anti-hepatitis B virus responses in rhesus

monkeys primed with a vectored measles virus and boosted with a single dose of hepatitis B surface antigen. Journal of virology, 2009. 83(17): p. 9013-9017.

20. Guy, B. and N. Jackson, Dengue vaccine: hypotheses to understand CYD-TDV-

induced protection. Nature Reviews Microbiology, 2015. 21. Brandler, S., et al., Pediatric measles vaccine expressing a dengue antigen

induces durable serotype-specific neutralizing antibodies to dengue virus. PLoS neglected tropical diseases, 2007. 1(3): p. e96.

22. Brandler, S., et al., A recombinant measles vaccine expressing chikungunya virus-

like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine, 2013. 31(36): p. 3718-3725.

23. Brandler, S., et al., Pediatric measles vaccine expressing a dengue tetravalent

antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine, 2010. 28(41): p. 6730-9.

24. Brandler, S. and F. Tangy, Recombinant vector derived from live attenuated

measles virus: potential for flavivirus vaccines. Comparative immunology, microbiology and infectious diseases, 2008. 31(2-3): p. 271-91.

Page 117: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

106

CHAPTER 5

SUMMARY AND FUTURE DIRECTIONS

Altogether, this work provides proof of principle for a novel vaccination platform

to induce neutralizing responses in protective range aganist Dengue virus infections,

while maintaining the protective measles vaccine phenotype. In chapter 2, I described

homologous and cross-immunity within the same subgroups of the flavivirus (DV2, DV4,

and DV1) and the related YFV. Cross-immunity between the various flavivirus can be

sufficiently construed in a host as a mean of generating balanced and a broad vaccine

against dengue virus and other closely related arthropod-borne viral pathogens. In chapter

3, I described the generation of a Measles-Dengue pediatric vaccine that induces

neutralizing immune responses against Measles, Dengue type 1,2,4 and YFV. This study

suggests a novel strategy for a pediatric anti-flavivirus vaccine to prime a population’s

immune response against various flavivirus infections. With a focus in immunity against

Dengue virus infections. I generated three recombinant measles expressing DV2 or 4

glycoproteins at high expression levels using a vaccine-equivalent measles genome; I

also described the homologous immunogenicity of single component vector and cocktails

of two vectors expressing DV2 and DV4 antigens. The neutralization potential of the

cocktail of recombinant measles vector was determined against DV1 and YFV. In

chapter 4, I documented that recombinant measles expressing DV4 PrM and full-length

major E protein generates neutralizing immune responses against Measles virus and a

weak anti-DV4 immune responses. This observation is important because it reflects a

correlation between antigenic exposure and neutralizing immune responses. Furthermore,

Page 118: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

107

since it also coincides with the lack of anti-DV2 immunity when using a full-length

antigen, it is arguable a consistent event for arthropod-borne flaviviruses or even related

agents like hepatitis C virus, that sequester their envelope proteins in an attempt to escape

native immunity. In sum, it may reflect a trend for flaviviral vaccine responses.

Additionally, I presented an alternative measurement of anti-flavivirus neutralizing

antibodies. I described the correlation between expressing the titer of neutralizing

antibodies by log neutralization index and the PRNT50. The log neutralization index

method depends on the direct measurements of the reduction of the viral titer as it relates

to non-relative serum such as pre-immune.

This project primarily depended upon the well-established measles reverse

genetic system[9,10], which has been used to generate many and different recombinants

and modified MVs[1, 2-6]. The availability of a full-length infectious cDNA encoding a

Moraten equivalent genome with engineered unique restriction sites for insertion of an

additional coding sequence in multiple loci in MV genome is practically convenient, for

this work insertion of foreign coding sequences in high level expressing locus proved

essential to obtain robust immune responses[4]. Immunogenicity studies presented this

dissertation depended on the availability of genetically modified measles susceptible

small animal model, the HuCD46Ge-IFNarKO mouse, which is the most reliable and

relevant small animal model for preclinical evaluation of MV vaccines.

This dissertation contributes to the field of Flavivirus vaccines development in

three clearly discernible ways. First, while previous studies have demonstrated cross

immune responses between different dengue serotypes[10,11], ours is the first study to

Page 119: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

108

show that these responses can be obtained using a recombinant vaccine, MV-Dengue

multivalent vaccine developed a homologous and heterologous neutralizing immune

responses against DV1, 2,4 and YFV. However, there have been limited investigations

of cross-protection between different subgroups. YFV and DVs belong to different sero-

complexes. In the present study, we demonstrated cross-immunity against YFV at

protective levels, which was induced by the recombinant Measles-Dengue 2 and 4

cocktail vaccine in the mice. Our results provide valuable information in understanding

the interaction between different flavivirus members, which further assist the

development of a novel multivalent vaccine against DVs serotypes, and multiple

flaviviruses such as YFV and JEV.

Second, and significantly, this dissertation identifies a promising complementary

anti-dengue immunization strategy based on a recombinant measles virus vaccine

cocktail to protect children in the age range from 2 to 9 years, whom may develop severe

forms of DV infection. Recently, the long-term follow-up results for the third year of the

phase III trial of the The Sanofi-Pasteur vaccine, Dengvaxia, conducted in Southeast Asia

(~10,000 children aged 2 to 14 years) result showed that vaccinees in the youngest age

group, younger than 5 years, had a substantially and significantly higher risk of

hospitalization for severing dengue disease than controls[7]. Importantly, seropositive

vaccine recipients had high and sustained antibody levels after the first dose of vaccine;

seronegative recipients had a lower peak of neutralizing antibodies by a factor of 10

times. The reduced efficacy in seronegative recipients especially younger age groups

suggested that the effectiveness of DV vaccine would mainly depend on the serostatus at

Page 120: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

109

the date of vaccination[]. In this dissertation, we proposed that a MV-Dengue pediatric

vaccine would induce broad anti-flaviral responses when administered to one-year infants

to prime their immune system. Protective flavivirus neutralization will be substantially

boosted upon administration of the subsequent chimeric Dengue vaccine. This approach

would close the current gap (younger than 9 years of age) and geographical indication

(only for hyperendemic countries), which makes the current dengue vaccine so

unpractical.

In a Third immense contribution to the field, this dissertation presents an idea for

a new generation of flavivirus vaccines which, may provide effective cross-protective

immunity against heterologous flavivirus infection in the endemic countries. Serological

cross-reactivity providing cross-protective immunity between related viruses has been

proven more than 200 years ago, as it was the immunological basis for the first human

vaccine against smallpox[14-16]. Indeed, whatever the nature of the attenuated virus used

as vaccine to eradicate smallpox (most probably camelpox or horsepox), cross-protective

responses proved safe and efficient. Cross protective flaviviral responses explain the

global distribution of mosquito-borne flaviviruses, for instance the practical absence of

yellow fever virus in Asia or the abysmal reduction of St Louis Encephalitis cases in the

USA after the introduction of West Nile virus. This study offers a platform to produce a

multivalent anti-flavivirus vaccine; the cocktail of recombinant vaccine may include

other recombinant measles expressing major E protein of other close flaviviruses

including YFV, JEV, and WNV. The cocktail of recombinant measles expressing

Page 121: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

110

multiple flavivirus antigens can be modified based on the co-circulation of the viruses

within different endemic countries.

In conclusion, this dissertation explores the generation of a recombinant measles-

dengue cocktail immunization to prime a broad, anti-flaviviral response in one-year-old

recipients as a complementary immunization strategy to enhance the immunogenicity and

efficiency of subsequent chimeric Dengue vaccine approaches. I sincerely hope that this

work will prove generative to the intelligent design of vaccines against DVs and other

flaviviruses.

Page 122: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

111

REFRENCES

1. Abe, Y., et al., Characteristics of viruses derived from nude mice with persistent

measles virus infection. Journal of virology, 2013. 87(8): p. 4170-5. 2. Brandler, S., et al., Pediatric measles vaccine expressing a dengue antigen

induces durable serotype-specific neutralizing antibodies to dengue virus. PLoS neglected tropical diseases, 2007. 1(3): p. e96.

3. Brandler, S., et al., Pediatric measles vaccine expressing a dengue tetravalent

antigen elicits neutralizing antibodies against all four dengue viruses. Vaccine, 2010. 28(41): p. 6730-9.

4. Brandler, S. and F. Tangy, Recombinant vector derived from live attenuated

measles virus: potential for flavivirus vaccines. Comparative immunology, microbiology and infectious diseases, 2008. 31(2-3): p. 271-91.

5. Lorin, C., et al., A paediatric vaccination vector based on live attenuated measles

vaccine. Therapie, 2005. 60(3): p. 227-33. 6. del Valle, J.R., et al., A vectored measles virus induces hepatitis B surface antigen

antibodies while protecting macaques against measles virus challenge. Journal of virology, 2007. 81(19): p. 10597-605.

7. ClinicalTrials.gov. Study of a novel tetravalent dengue vaccine in healthy children

aged 2 to 14 years in Asia. 2015 [cited 2016 10-14-2016]; Available from: http://clinicaltrials.gov/show/NCT01373281.

8. Guy, B. and N. Jackson, Dengue vaccine: hypotheses to understand CYD-TDV-

induced protection. Nature Reviews Microbiology, 2015. 9. Parks, C.L., et al., Enhanced measles virus cDNA rescue and gene expression

after heat shock. Journal of virology, 1999. 73(5): p. 3560-3566. 10. Radecke, F., et al., Rescue of measles viruses from cloned DNA. The EMBO

journal, 1995. 14(23): p. 5773. 11. Vaughn, D.W., et al., Dengue viremia titer, antibody response pattern, and virus

serotype correlate with disease severity. Journal of Infectious Diseases, 2000. 181(1): p. 2-9.

12. Rothman, A.L., Dengue: defining protective versus pathologic immunity. The

Journal of clinical investigation, 2004. 113(7): p. 946-951.

Page 123: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

112

13. Dejnirattisai, W., et al., Cross-reacting antibodies enhance dengue virus infection

in humans. Science, 2010. 328(5979): p. 745-748. 14. Lobigs, M. and M.S. Diamond, Feasibility of cross-protective vaccination against

flaviviruses of the Japanese encephalitis serocomplex. Expert review of vaccines, 2012. 11(2): p. 177-187.

15. Mansfield, K.L., et al., Flavivirus-induced antibody cross-reactivity. Journal of

general Virology, 2011. 92(12): p. 2821-2829. 16. Li, J., et al., Cross-protection induced by Japanese encephalitis vaccines against

different genotypes of Dengue viruses in mice. Scientific reports, 2016. 6.

Page 124: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

113

COMPREHENSIVE LIST OF REFERENCES Abe, Y., K. Hashimoto, et al. (2013). "Characteristics of viruses derived from nude mice

with persistent measles virus infection." Journal of virology 87(8): 4170-4175. Amar-Singh, H., M.-T. Koh, et al. (2013). "Safety and immunogenicity of a tetravalent

dengue vaccine in healthy children aged 2–11 years in Malaysia: A randomized, placebo-controlled, Phase III study." Vaccine 31(49): 5814-5821.

Anderson, K. B., R. V. Gibbons, et al. (2011). "Preexisting Japanese encephalitis virus

neutralizing antibodies and increased symptomatic dengue illness in a school-based cohort in Thailand." PLoS Negl Trop Dis 5(10): e1311.

Bartenschlager, R. and S. Miller (2008). "Molecular aspects of Dengue virus replication."

Future microbiology 3(2): 155-165. Beckett, C. G., J. Tjaden, et al. (2011). "Evaluation of a prototype dengue-1 DNA

vaccine in a Phase 1 clinical trial." Vaccine 29(5): 960-968. Beltramello, M., K. L. Williams, et al. (2010). "The human immune response to Dengue

virus is dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity." Cell host & microbe 8(3): 271-283.

Blackley, S., Z. Kou, et al. (2007). "Primary human splenic macrophages, but not T or B

cells, are the principal target cells for dengue virus infection in vitro." Journal of virology 81(24): 13325-13334.

Boonnak, K., B. M. Slike, et al. (2008). "Role of dendritic cells in antibody-dependent

enhancement of dengue virus infection." Journal of virology 82(8): 3939-3951. Brandler, S., M. Lucas-Hourani, et al. (2007). "Pediatric measles vaccine expressing a

dengue antigen induces durable serotype-specific neutralizing antibodies to dengue virus." PLoS neglected tropical diseases 1(3): e96.

Brandler, S., C. Ruffié, et al. (2013). "A recombinant measles vaccine expressing

chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus." Vaccine 31(36): 3718-3725.

Brandler, S., C. Ruffie, et al. (2010). "Pediatric measles vaccine expressing a dengue

tetravalent antigen elicits neutralizing antibodies against all four dengue viruses." Vaccine 28(41): 6730-6739.

Page 125: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

114

Brandler, S. and F. Tangy (2008). "Recombinant vector derived from live attenuated measles virus: potential for flavivirus vaccines." Comparative immunology, microbiology and infectious diseases 31(2-3): 271-291.

Bredenbeek, P. J., E. A. Kooi, et al. (2003). "A stable full-length yellow fever virus cDNA clone and the role of conserved RNA elements in flavivirus replication." Journal of general virology 84(5): 1261-1268.

Bukowski, J. F., I. Kurane, et al. (1989). "Dengue virus-specific cross-reactive CD8+

human cytotoxic T lymphocytes." Journal of virology 63(12): 5086-5091. Butthep, P., S. Chunhakan, et al. (2006). "Elevated soluble thrombomodulin in the febrile

stage related to patients at risk for dengue shock syndrome." The Pediatric infectious disease journal 25(10): 894-897.

Capeding, M. R., N. H. Tran, et al. (2014). "Clinical efficacy and safety of a novel

tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial." The Lancet 384(9951): 1358-1365.

Carlos, C. C., K. Oishi, et al. (2005). "Comparison of clinical features and hematologic

abnormalities between dengue fever and dengue hemorrhagic fever among children in the Philippines." The American journal of tropical medicine and hygiene 73(2): 435-440.

Chen, Y., T. Maguire, et al. (1997). "Dengue virus infectivity depends on envelope

protein binding to target cell heparan sulfate." Nature medicine 3(8): 866-871. Churdboonchart, V., N. Bhamarapravati, et al. (1991). "Antibodies against dengue viral

proteins in primary and secondary dengue hemorrhagic fever." The American journal of tropical medicine and hygiene 44(5): 481-493.

ClinicalTrails.gov. (2015). "Study of ChimeriVax™ tetravalent dengue vaccine in

healthy subjects." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT00875524.

ClinicalTrials.gov. (2014). "TDENV PIV and LAV dengue prime-boost strategy."

Retrieved 10-14-2016, 2016, from https://clinicaltrials.gov/show/NCT02239614. ClinicalTrials.gov. (2015). "Evaluation of the safety and efficacy of a single dose of a

dengue vaccine (TV005) in healthy adults." Retrieved 10-14-2016, 2016, from https://clinicaltrials.gov/show/NCT02317900.

ClinicalTrials.gov. (2015). "Phase 1b study investigating safety & immunogenicity of

DENVax given intradermally by needle or needle free PharmaJet injector." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01765426.

Page 126: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

115

ClinicalTrials.gov. (2015). "Phase II trial to evaluate safety and immunogenicity of a

dengue 1,2,3,4 (attenuated) vaccine TV003/TV005." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01696422.

ClinicalTrials.gov. (2015). "Safety and immunogenicity of different schedules of

Takeda's tetravalent dengue vaccine candidate (TDV) in healthy participants." Retrieved 10-14-2016, 2016, from https://clinicaltrials.gov/show/NCT02302066.

ClinicalTrials.gov. (2015). "Safety and immunogenicity study to assess DENVax, a live

attenuated tetravalent vaccine for prevention of dengue ever." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01224639.

ClinicalTrials.gov. (2015). "Safety study of a dengue virus DNA vaccine." Retrieved

10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT00290147. ClinicalTrials.gov. (2015). "Study of a dengue vaccine (V180) in healthy adults (V180-

001." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01477580.

ClinicalTrials.gov. (2015). "Study of a novel tetravalent dengue vaccine in healthy

children aged 2 to 14 years in Asia." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01373281.

ClinicalTrials.gov. (2015). "A two-dose primary vaccination study of a tetravalent

dengue virus purified inactivated vaccine vs. placebo in healthy adults." Retrieved 10-14-2016, 2016, from http://clinicaltrials.gov/show/NCT01666652.

Coller, B.-A. G., D. E. Clements, et al. (2011). "The development of recombinant subunit

envelope-based vaccines to protect against dengue virus induced disease." Vaccine 29(42): 7267-7275.

Combredet, C., V. Labrousse, et al. (2003). "A molecularly cloned Schwarz strain of

measles virus vaccine induces strong immune responses in macaques and transgenic mice." Journal of virology 77(21): 11546-11554.

Dejnirattisai, W., A. Jumnainsong, et al. (2010). "Cross-reacting antibodies enhance

dengue virus infection in humans." Science 328(5979): 745-748. del Valle, J. R., P. Devaux, et al. (2007). "A vectored measles virus induces hepatitis B

surface antigen antibodies while protecting macaques against measles virus challenge." Journal of virology 81(19): 10597-10605.

Page 127: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

116

Delpeut, S., R. S. Noyce, et al. (2012). "Host factors and measles virus replication." Current opinion in virology 2(6): 773-783.

Despres, P., C. Combredet, et al. (2005). "Live measles vaccine expressing the secreted

form of the West Nile virus envelope glycoprotein protects against West Nile virus encephalitis." The Journal of infectious diseases 191(2): 207-214.

Durbin, A. P., R. A. Karron, et al. (2001). "Attenuation and immunogenicity in humans

of a live dengue virus type-4 vaccine candidate with a 30 nucleotide deletion in its 3'-untranslated region." The American journal of tropical medicine and hygiene 65(5): 405-413.

Durbin, A. P., J. McArthur, et al. (2006). "The live attenuated dengue serotype 1 vaccine

rDEN1Δ30 is safe and highly immunogenic in healthy adult volunteers." Human vaccines 2(4): 167-173.

Durbin, A. P., S. S. Whitehead, et al. (2005). "rDEN4Δ30, a live attenuated dengue

virus type 4 vaccine candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers." Journal of Infectious Diseases 191(5): 710-718.

Egloff, M. P., D. Benarroch, et al. (2002). "An RNA cap (nucleoside-2'-O-)-

methyltransferase in the flavivirus RNA polymerase NS5: crystal structure and functional characterization." The EMBO journal 21(11): 2757-2768.

Erlenhoefer, C., W. J. Wurzer, et al. (2001). "CD150 (SLAM) is a receptor for measles

virus but is not involved in viral contact-mediated proliferation inhibition." Journal of virology 75(10): 4499-4505.

Foster, J. E., S. N. Bennett, et al. (2004). "Phylogeography and molecular evolution of

dengue 2 in the Caribbean basin, 1981–2000." Virology 324(1): 48-59. Griffin, D. E. and M. B. Oldstone (2008). Measles: history and basic biology, Springer

Science & Business Media. Gubler, D. J. (1998). "Dengue and dengue hemorrhagic fever." Clinical microbiology

reviews 11(3): 480-496. Gubler, D. J. (2002). "Epidemic dengue/dengue hemorrhagic fever as a public health,

social and economic problem in the 21st century." Trends in microbiology 10(2): 100-103.

Guerbois, M., A. Moris, et al. (2009). "Live attenuated measles vaccine expressing HIV-1

Gag virus like particles covered with gp160ΔV1V2 is strongly immunogenic." Virology 388(1): 191-203.

Page 128: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

117

Guy, B., B. Barrere, et al. (2011). "From research to phase III: preclinical, industrial and

clinical development of the Sanofi Pasteur tetravalent dengue vaccine." Vaccine 29(42): 7229-7241.

Guy, B., O. Briand, et al. (2015). "Development of the Sanofi Pasteur tetravalent dengue

vaccine: One more step forward." Vaccine 33(50): 7100-7111. Guy, B., F. Guirakhoo, et al. (2010). "Preclinical and clinical development of YFV 17D-

based chimeric vaccines against dengue, West Nile and Japanese encephalitis viruses." Vaccine 28(3): 632-649.

Guy, B. and N. Jackson (2015). "Dengue vaccine: hypotheses to understand CYD-TDV-

induced protection." Nature Reviews Microbiology. Guzman, M. a. G., G. Kouri, et al. (2002). "Effect of age on outcome of secondary

dengue 2 infections." International journal of infectious diseases 6(2): 118-124. Guzman, M. G., G. Kouri, et al. (2000). "Epidemiologic studies on Dengue in Santiago

de Cuba, 1997." American journal of epidemiology 152(9): 793-799. Guzman, M. G., A. Alvarez, et al. (2012). "Epidemiological studies on dengue virus type

3 in Playa municipality, Havana, Cuba, 2001–2002." International journal of infectious diseases 16(3): e198-e203.

Guzman, M. G., S. B. Halstead, et al. (2010). "Dengue: a continuing global threat."

Nature Reviews Microbiology 8: S7-S16. Guzman, M. G. and E. Harris (2015). "Dengue." The Lancet 385(9966): 453-465. Hadinegoro, S. R., J. L. Arredondo-García, et al. (2015). "Efficacy and long-term

safety of a dengue vaccine in regions of endemic disease." New England Journal of Medicine 373(13): 1195-1206.

Halstead, S. B. (1970). "Observations related to pathogensis of dengue hemorrhagic

fever. VI. Hypotheses and discussion." The Yale journal of biology and medicine 42(5): 350-362.

Halstead, S. B. and P. Simasthien (1970). "Observations related to the pathogenesis of

dengue hemorrhagic fever. II. Antigenic and biologic properties of dengue viruses and their association with disease response in the host." The Yale journal of biology and medicine 42(5): 276.

Hardy, J. (1988). Susceptibility of vector Mosquitoes. The arboviruses: Epidemiology

and ecology. T. Monath. Boca Raton, FLA, CRC Press: 87-126.

Page 129: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

118

Henchal, E. A. and J. R. Putnak (1990). "The dengue viruses." Clinical microbiology

reviews 3(4): 376-396. Ho, L. J., J. J. Wang, et al. (2001). "Infection of human dendritic cells by dengue virus

causes cell maturation and cytokine production." Journal of immunology 166(3): 1499-1506.

Huerre, M. R., N. T. Lan, et al. (2001). "Liver histopathology and biological correlates in five cases of fatal dengue fever in Vietnamese children." Virchows Archiv : an international journal of pathology 438(2): 107-115.

Huerta, V., G. Chinea, et al. (2008). "Characterization of the interaction of domain III of

the envelope protein of dengue virus with putative receptors from CHO cells." Virus research 137(2): 225-234.

Institute, B. (July 21, 2016). "Phase III Trial to Evaluate Efficacy and Safety of a

Tetravalent Dengue Vaccine". Retrieved 08/08/2016, 2016, from https://clinicaltrials.gov/ct2/show/NCT02406729.

Jenkins, G. M., M. Pagel, et al. (2001). "Evolution of base composition and codon usage

bias in the genus Flavivirus." Journal of molecular evolution 52(4): 383-390. Jessie, K., M. Y. Fong, et al. (2004). "Localization of dengue virus in naturally infected

human tissues, by immunohistochemistry and in situ hybridization." The Journal of infectious diseases 189(8): 1411-1418.

Karber, G. (1931). "50% end-point calculation." Arch Exp Pathol Pharmak 162: 480-483. Kaufmann, B. and M. G. Rossmann (2011). "Molecular mechanisms involved in the early

steps of flavivirus cell entry." Microbes and infection / Institut Pasteur 13(1): 1-9. Kliks, S. C., S. Nimmanitya, et al. (1988). "Evidence that maternal dengue antibodies are

important in the development of dengue hemorrhagic fever in infants." The American journal of tropical medicine and hygiene 38(2): 411-419.

Kliks, S. C., A. Nisalak, et al. (1989). "Antibody-dependent enhancement of dengue virus

growth in human monocytes as a risk factor for dengue hemorrhagic fever." The American journal of tropical medicine and hygiene 40(4): 444-451.

Kosasih, H., H. Yusuf, et al. (2006). "Report of four volunteers with primary, secondary

and tertiary dengue infections during a prospective cohort study." Dengue Bulletin 30: 87.

Page 130: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

119

Kuno, G., G.-J. J. Chang, et al. (1998). "Phylogeny of the genus Flavivirus." Journal of virology 72(1): 73-83.

Kurane, I., A. Meager, et al. (1989). "Dengue virus-specific human T cell clones.

Serotype crossreactive proliferation, interferon gamma production, and cytotoxic activity." The Journal of experimental medicine 170(3): 763-775.

Kwan, W. H., A. M. Helt, et al. (2005). "Dendritic cell precursors are permissive to

dengue virus and human immunodeficiency virus infection." Journal of virology 79(12): 7291-7299.

Lanciotti, R. S., D. J. Gubler, et al. (1997). "Molecular evolution and phylogeny of dengue-4 viruses." Journal of General Virology 78(9): 2279-2284.

Lei, H. Y., T. M. Yeh, et al. (2001). "Immunopathogenesis of dengue virus infection."

Journal of biomedical science 8(5): 377-388. Li, J., N. Gao, et al. (2016). "Cross-protection induced by Japanese encephalitis vaccines

against different genotypes of Dengue viruses in mice." Scientific reports 6. Li, L., S.-M. Lok, et al. (2008). "The flavivirus precursor membrane-envelope protein

complex: structure and maturation." Science 319(5871): 1830-1834. Lisova, O., F. Hardy, et al. (2007). "Mapping to completeness and transplantation of a

group-specific, discontinuous, neutralizing epitope in the envelope protein of dengue virus." Journal of General Virology 88(9): 2387-2397.

Lobigs, M. (1993). "Flavivirus premembrane protein cleavage and spike heterodimer

secretion require the function of the viral proteinase NS3." Proceedings of the National Academy of Sciences of the United States of America 90(13): 6218-6222.

Lobigs, M. and M. S. Diamond (2012). "Feasibility of cross-protective vaccination

against flaviviruses of the Japanese encephalitis serocomplex." Expert review of vaccines 11(2): 177-187.

Lorin, C., C. Combredet, et al. (2005). "A paediatric vaccination vector based on live

attenuated measles vaccine." Therapie 60(3): 227-233. Lorin, C., L. Mollet, et al. (2004). "A single injection of recombinant measles virus

vaccines expressing human immunodeficiency virus (HIV) type 1 clade B envelope glycoproteins induces neutralizing antibodies and cellular immune responses to HIV." Journal of virology 78(1): 146-157.

Page 131: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

120

Lozach, P. Y., L. Burleigh, et al. (2005). "Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN)-mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals." The Journal of biological chemistry 280(25): 23698-23708.

Lund, J. M., L. Alexopoulou, et al. (2004). "Recognition of single-stranded RNA viruses

by Toll-like receptor 7." Proceedings of the National Academy of Sciences of the United States of America 101(15): 5598-5603.

Luo, D., T. Xu, et al. (2008). "Crystal structure of the NS3 protease-helicase from dengue

virus." Journal of virology 82(1): 173-183. Mangada, M. M. and A. L. Rothman (2005). "Altered cytokine responses of dengue-

specific CD4+ T cells to heterologous serotypes." Journal of immunology 175(4): 2676-2683.

Mansfield, K. L., D. L. Horton, et al. (2011). "Flavivirus-induced antibody cross-

reactivity." Journal of general Virology 92(12): 2821-2829. Martina, B. E., P. Koraka, et al. (2009). "Dengue virus pathogenesis: an integrated view."

Clinical microbiology reviews 22(4): 564-581. Mason, R. A., N. M. Tauraso, et al. (1973). "Yellow fever vaccine: direct challenge of

monkeys given graded doses of 17D vaccine." Applied microbiology 25(4): 539-544.

Montoya, M., L. Gresh, et al. (2013). "Symptomatic versus inapparent outcome in repeat

dengue virus infections is influenced by the time interval between infections and study year." PLoS Negl Trop Dis 7(8): e2357.

Morrison, J., S. Aguirre, et al. (2012). "Innate immunity evasion by Dengue virus."

Viruses 4(3): 397-413. Mrkic, B., J. Pavlovic, et al. (1998). "Measles virus spread and pathogenesis in

genetically modified mice." Journal of virology 72(9): 7420-7427. Mrkic, B., J. Pavlovic, et al. (1998). "Measles virus spread and pathogenesis in

genetically modified mice." Journal of virology 72(9): 7420-7427. Muhlebach, M. D., M. Mateo, et al. (2011). "Adherens junction protein nectin-4 is the

epithelial receptor for measles virus." Nature 480(7378): 530-533. Munoz-Jordan, J. L., G. G. Sanchez-Burgos, et al. (2003). "Inhibition of interferon

signaling by dengue virus." Proceedings of the National Academy of Sciences of the United States of America 100(24): 14333-14338.

Page 132: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

121

Naniche, D., G. Varior-Krishnan, et al. (1993). "Human membrane cofactor protein

(CD46) acts as a cellular receptor for measles virus." Journal of virology 67(10): 6025-6032.

Navarro-Sanchez, E., P. Despres, et al. (2005). "Innate immune responses to dengue

virus." Archives of medical research 36(5): 425-435. Nimmannitya, S. (1987). "Clinical spectrum and management of dengue haemorrhagic

fever." The Southeast Asian journal of tropical medicine and public health 18(3): 392-397.

OhAinle, M., A. Balmaseda, et al. (2011). "Dynamics of dengue disease severity determined by the interplay between viral genetics and serotype-specific immunity." Science translational medicine 3(114): 114ra128-114ra128.

Ono, N., H. Tatsuo, et al. (2001). "Measles viruses on throat swabs from measles patients

use signaling lymphocytic activation molecule (CDw150) but not CD46 as a cellular receptor." Journal of virology 75(9): 4399-4401.

Organization, W. H. (2012). "Dengue and severe dengue." WHO Fact sheet No117

(Updated September 2014) http://www. who. int/mediacentre/factsheets/fs117/en. Organization, W. H. (2012). "Global strategy for dengue prevention and control 2012-

2020." Osorio, J. E., C. Y.-H. Huang, et al. (2011). "Development of DENVax: a chimeric

dengue-2 PDK-53-based tetravalent vaccine for protection against dengue fever." Vaccine 29(42): 7251-7260.

Parks, C. L., R. A. Lerch, et al. (1999). "Enhanced measles virus cDNA rescue and gene

expression after heat shock." Journal of virology 73(5): 3560-3566. Perera, R. and R. J. Kuhn (2008). "Structural proteomics of dengue virus." Current

opinion in microbiology 11(4): 369-377. PRICE, W. H. (1968). "Sequential immunization as a vaccination procedure against

dengue viruses." American journal of epidemiology 88(3): 392-397. PRICE, W. H. and I. S. THIND (1971). "Protection against West Nile virus induced by a

previous injection with dengue virus." American journal of epidemiology 94(6): 596-607.

Radecke, F. and M. Billeter (1995). Appendix: measles virus antigenome and protein

consensus sequences. Measles Virus, Springer: 181-192.

Page 133: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

122

Radecke, F., P. Spielhofer, et al. (1995). "Rescue of measles viruses from cloned DNA."

The EMBO journal 14(23): 5773-5784. Ramsauer, K., M. Schwameis, et al. (2015). "Immunogenicity, safety, and tolerability of

a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial." The Lancet infectious diseases 15(5): 519-527.

Reich, N. G., S. Shrestha, et al. (2013). "Interactions between serotypes of dengue

highlight epidemiological impact of cross-immunity." Journal of The Royal Society Interface 10(86): 20130414.

Reyes-del Valle, J., C. de la Fuente, et al. (2012). "Broadly neutralizing immune responses against hepatitis C virus induced by vectored measles viruses and a recombinant envelope protein booster." Journal of virology 86(21): 11558-11566.

Reyes-del Valle, J., G. Hodge, et al. (2009). "Protective anti-hepatitis B virus responses

in rhesus monkeys primed with a vectored measles virus and boosted with a single dose of hepatitis B surface antigen." Journal of virology 83(17): 9013-9017.

Rodrigo, W. S. I., O. K. Block, et al. (2009). "Dengue virus neutralization is modulated

by IgG antibody subclass and Fcγ receptor subtype." Virology 394(2): 175-182. Rothman, A. L. (2004). "Dengue: defining protective versus pathologic immunity." The

Journal of clinical investigation 113(7): 946-951. Rothman, A. L. (2011). "Immunity to dengue virus: a tale of original antigenic sin and

tropical cytokine storms." Nature reviews. Immunology 11(8): 532-543. Sabin, A. B. (1952). "Research on dengue during World War II." American journal of

tropical medicine and hygiene 1(1): 30-50. SANGKAWIBHA, N., S. ROJANASUPHOT, et al. (1984). "Risk factors in dengue

shock syndrome: a prospective epidemiologic study in Rayong, Thailand I. The 1980 outbreak." American journal of epidemiology 120(5): 653-669.

Schieffelin, J. S., J. M. Costin, et al. (2010). "Neutralizing and non-neutralizing

monoclonal antibodies against dengue virus E protein derived from a naturally infected patient." Virology journal 7: 28.

Schlesinger, R. W., I. Gordon, et al. (1956). "Clinical and serologic response of man to

immunization with attenuated dengue and yellow fever viruses." The Journal of Immunology 77(5): 352-364.

Page 134: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

123

Schwartz, L. M., M. E. Halloran, et al. (2015). "The dengue vaccine pipeline:

Implications for the future of dengue control." Vaccine 33(29): 3293-3298. Shaio, M. F., F. Y. Chang, et al. (1992). "Complement pathway activity in serum from

patients with classical dengue fever." Transactions of the Royal Society of Tropical Medicine and Hygiene 86(6): 672-675.

Sidhu, M. S., J. Chan, et al. (1995). "Rescue of synthetic measles virus minireplicons:

measles genomic termini direct efficient expression and propagation of a reporter gene." Virology 208(2): 800-807.

Sin Leo, Y., A. Wilder-Smith, et al. (2012). "Immunogenicity and safety of recombinant

tetravalent dengue vaccine (CYD-TDV) in individuals aged 2–45 years: Phase II randomized controlled trial in Singapore." Human vaccines & immunotherapeutics 8(9): 1259-1271.

Stefan Flasche1*, M. J., Isabel Rodríguez-Barraquer 2*, Laurent Coudeville3*, Mario

Recker4*,, G. M. Katia Koelle5*, Tom Hladish7*, Alex Perkins8*, Ilaria Dorigatti9, et al. (2016). Comparative modelling of dengue vaccine public health impact, World Health Organization.

Stiasny, K., R. Fritz, et al. (2011). "Molecular mechanisms of flavivirus membrane

fusion." Amino acids 41(5): 1159-1163. Stiasny, K. and F. X. Heinz (2006). "Flavivirus membrane fusion." The Journal of

general virology 87(Pt 10): 2755-2766. Takeda. (2016). "Safety and Immunogenicity With Two Different Serotype 2 Potencies

of Tetravalent Dengue Vaccine (TDV) in Adults in Singapore." Retrieved 08/08/2016, 2016, from https://clinicaltrials.gov/ct2/show/study/NCT02425098?term=dengue+and+takeda&rank=4.

Tangy, F. and H. Y. Naim (2005). "Live attenuated measles vaccine as a potential

multivalent pediatric vaccination vector." Viral immunology 18(2): 317-326. Tatsuo, H., N. Ono, et al. (2001). "Morbilliviruses use signaling lymphocyte activation

molecules (CD150) as cellular receptors." Journal of virology 75(13): 5842-5850. Thullier, P., C. Demangel, et al. (2001). "Mapping of a dengue virus neutralizing epitope

critical for the infectivity of all serotypes: insight into the neutralization mechanism." Journal of General Virology 82(8): 1885-1892.

Page 135: Vaccination Strategy To Protect Against Flavivirus ...€¦ · solution to complement the current vaccine: a recombinant measles vaccine vectoring dengue protective antigens to be

124

Valdes, K., M. Alvarez, et al. (2000). "Human Dengue antibodies against structural and

nonstructural proteins." Clinical and diagnostic laboratory immunology 7(5): 856-857.

Vaughn, D. W., S. Green, et al. (2000). "Dengue viremia titer, antibody response pattern,

and virus serotype correlate with disease severity." The Journal of infectious diseases 181(1): 2-9.

Villar, L., G. H. Dayan, et al. (2015). "Efficacy of a tetravalent dengue vaccine in

children in Latin America." New England Journal of Medicine 372(2): 113-123. Wahala, W. M., A. A. Kraus, et al. (2009). "Dengue virus neutralization by human

immune sera: role of envelope protein domain III-reactive antibody." Virology 392(1): 103-113.

Wang, E., H. Ni, et al. (2000). "Evolutionary relationships of endemic/epidemic and sylvatic dengue viruses." Journal of virology 74(7): 3227-3234.

Watanabe, S., Y. Shirogane, et al. (2013). "Mutant fusion proteins with enhanced fusion

activity promote measles virus spread in human neuronal cells and brains of suckling hamsters." Journal of virology 87(5): 2648-2659.

Whitehead, S. S., J. E. Blaney, et al. (2007). "Prospects for a dengue virus vaccine."

Nature Reviews Microbiology 5(7): 518-528. Wilder-Smith, A. and D. J. Gubler (2015). "Dengue vaccines at a crossroad." Science

350(6261): 626-627. Wright, P. F., A. P. Durbin, et al. (2009). "Phase 1 trial of the dengue virus type 4 vaccine

candidate rDEN4Δ30-4995 in healthy adult volunteers." The American journal of tropical medicine and hygiene 81(5): 834-841.

Wu, S. J., G. Grouard-Vogel, et al. (2000). "Human skin Langerhans cells are targets of

dengue virus infection." Nature medicine 6(7): 816-820. Yauch, L. E., R. M. Zellweger, et al. (2009). "A protective role for dengue virus-specific

CD8+ T cells." Journal of immunology 182(8): 4865-4873. Zhang, Y., J. Corver, et al. (2003). "Structures of immature flavivirus particles." The

EMBO journal 22(11): 2604-2613. Zhou, Y., D. Ray, et al. (2007). "Structure and function of flavivirus NS5

methyltransferase." Journal of virology 81(8): 3891-3903.