engineering dna vaccines that include plant virus coat proteins

16

Click here to load reader

Upload: freda-k

Post on 15-Apr-2017

215 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

This article was downloaded by: [University of Louisville]On: 20 December 2014, At: 18:47Publisher: Taylor & FrancisInforma Ltd Registered in England and Wales Registered Number: 1072954Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH,UK

Biotechnology and GeneticEngineering ReviewsPublication details, including instructions forauthors and subscription information:http://www.tandfonline.com/loi/tbgr20

Engineering DNA Vaccinesthat Include Plant Virus CoatProteinsNatalia Savelyeva a , Delin Zhu a & Freda K.Stevenson aa Molecular Immunology Group, Cancer SciencesDivision, Southampton University Hospitals Trust,Tremona Road , Southampton , SOW 6YD , UKPublished online: 15 Apr 2013.

To cite this article: Natalia Savelyeva , Delin Zhu & Freda K. Stevenson (2003)Engineering DNA Vaccines that Include Plant Virus Coat Proteins, Biotechnology andGenetic Engineering Reviews, 20:1, 101-116, DOI: 10.1080/02648725.2003.10648039

To link to this article: http://dx.doi.org/10.1080/02648725.2003.10648039

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all theinformation (the “Content”) contained in the publications on our platform.However, Taylor & Francis, our agents, and our licensors make norepresentations or warranties whatsoever as to the accuracy, completeness,or suitability for any purpose of the Content. Any opinions and viewsexpressed in this publication are the opinions and views of the authors, andare not the views of or endorsed by Taylor & Francis. The accuracy of theContent should not be relied upon and should be independently verified withprimary sources of information. Taylor and Francis shall not be liable for anylosses, actions, claims, proceedings, demands, costs, expenses, damages,and other liabilities whatsoever or howsoever caused arising directly orindirectly in connection with, in relation to or arising out of the use of theContent.

Page 2: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

This article may be used for research, teaching, and private study purposes.Any substantial or systematic reproduction, redistribution, reselling, loan,sub-licensing, systematic supply, or distribution in any form to anyone isexpressly forbidden. Terms & Conditions of access and use can be found athttp://www.tandfonline.com/page/terms-and-conditions

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 3: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

5Engineering DNA Vaccines that IncludePlant Virus Coat Proteins

NATALIA SAVELYEVA3, DELIN ZHU AND FREDA K. STEVENSON

Molecular /rI'l111uno[ogy Group, Cancer Sciences Division, Southam/Jton UniversityHospitals Trust, Trel1101Za Road, Southal11ptOll, 8016 6YD, UK

Introduction

There is growing interest among researchers in the various strategies for vaccinationagainst cancer. One prolnising approach is the use of naked plasmid DNA for antigendelivery (Tang etai., 1992). An advantage ofDNA vaccination is the ability to inducehumoral, as well as cellular immune responses to control tumours. DNA vaccines canbe made easily using standard DNA recombination technology, thus allowing manycandidate antigens to be tested rapidly.

A DNA vaccine combines two functional units; one encodes the antigen of interest,and the other acts as an adjuvant. A cassette capable of driving gene expression ineukaryotic cells is inserted into a plasmid. This cassette usually includes a CMVprolnoter/enhancer and a terminator/polyadenylation signal (Figure 5.1). Upondelivery of the bacterial plasmid DNA into muscle or skin~ the gene is expressed,producing the antigen directly in transfected cells. The adjuvant effect comes fromiInlnunostiJnulatory DNA sequences (ISSs)~ i.e. unmethylated CpG motifs withinparticular flanking sequences (Krieg et at., 1995). These motifs are more frequent inbacterial than in eukaryotic DNA, and act as so-called 'danger' signals of bacterialinvasion. ISS are recognized by the cells of the ilTItnUne system, such as dendriticcells, B cells, and natural killer cells (review: Krieg, 2002). This leads to release ofinflammatory cytokines, such as IFN-y, IL-12 and IL-18, chemokine secretion, andupregulation of co-stitnulatory tTIolecules on APes. The exact sequences and fre­quency of ISS within a plasnlid DNA backbone vary for different species. TLR9, aJnetnber of the TLR-receptor family, has been identified as a pattern recognition

*To whOln correspondence Inay be addressed: ([email protected])

Abbreviations: APe. antigen-presenting cell; BCR~ B cell receptor; CMV.. cytolnegalovirus; CPt coalprotein~ DC. dendritic cells; DR. death receptor; FrC, fragment C; Id, idiotypic determinants; IFN,interferon; Ig, immunoglobulin: IL.. jnterJeukin: MAb, monoclonal antibody; MHC, Inain histocompatibil­ity complex~PVX, potato virus X: TA, tumour antigen~TCR~T cell receptor; Th. T helper; TLR.. ToU-likercceptor~TNF, tumour necrosis factor; TRAIL, TNF-related apoprosis-inducing ligand; TWEAK, TNF-likeweak jnducer of apoptosis.

8iotecJlIlalogy (lIul (ienetic E.i,g;1Jeerillg Rt!\';ews - Vol. 20, December 20030264-8725/03/20/101-114 $20.00 + $0.00 © Intercept Ltd. PO Box 716. Andover, Hamp~hirc, SPIO 1YO. UK

101

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 4: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

102 N. SAVELYEVA etal.

DNA Cytokine

vaccine release IFNv a,1} }IL-12 IFN-y

JL-18

/ JIt ~ ~ISS

Antigen-presentingMuscle/skin -------.

Antigen cell

cells transfer

/ 1 \antibody CD4+ ceUs CD8+ cells

Th1>Th2

Figure 5.1. Induction of the immune response by DNA vaccines. ISSs containing CpG lnotifs in theplasmid backbone stinlulate the production of pro~jnflalnmatorycytokines by cells of the innate hnmuneSystCID through interaction with TLR9. Transfection of muscle/skin and dendritic cells leads to antigenexpression~ \vith a subsequent activation of CD4+ and CD8+ TceHs and antibody.

receptor for uilinethylated CpO motifs (Hemlni et al., 2000). Activation of TLR-9leads to induction of an NF-ICB signal transduction pathway (Hetnnli ef al., 2000).

Plaslnid DNA is usually delivered by intramuscular injection, or an intradermalinjection using a gene gun. Myocytes or keratinocytes are transfected t and these cellsthen act as a major source of antigen. It is possible that APe can also be transfecteddirectly (Condon et aI., 1996). In addition, APe can acquire soluble antigent or takeup ceJl debt; s, which have occurred as a result of apoptotic or necrotic death oftransfected cells in the site of injection (Fu et al., 1997). These APe migrate to thedraining lymph nodes, where they prinle na"ive T ceUs (Bot et aI., 2000). All arms ofthe hnmune response can be induced t including antibody, CD4+ T cells and cytotoxicCD8+ T cells. Intramuscular injection generally induces a Thl type of immuneresponse, which is associated with activation ofceJlular inlmunity, whereas the genegun method favours a Th2 type response with induction of hlunoral responses(Feltquate et al., 1997). However, the nature of the antigen can also influence the typeof helper response induced during DNA vaccination.

For DNA vaccination against cancer, our strategy is to use genes that code for TA.We have focused on Id, a specific TA expressed by B cell malignancies. Ids areencoded by variable regions of heavy and light chains of Ig genes t and are unique foreach B cell. Rearrangelnent of V, D, and J gene segments of the heavy chain, and Vand J segments of the light chain during B cell differentiation leads to the expressionof an immunoglobulin of a single specificity. During antigen-induced maturation of

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 5: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include plant virus coat proteins 103

a B cell, further diversity of 19 is achieved by somatic hypermulation. Ids provide aclear tumour-specific target for immune attack against B cell tumours. Vaccinationwith purified idiotypic Ig protein induces specific protective antibody-mediatedimmunity in a number of nlouse Jymphonla tnodels (CampbeU et al., 1987; George etaI., 1987). This principle of idiotypic vaccination is also applicable to patients, asdenlonstrated by the results of recent clinical trials (Hsu et aL t 1997). However, awide clinical application of Id protein vaccines is hindered by the need to prepareprotein vaccines for each patient individually. DNA vaccination offers a moreconvenient alternative approach.

In a number of Illodels of infectious diseases, a simple DNA delivery of pathogen­derived antigen alone can induce potent immune responses. However, for B celltumours, DNA vaccination with Id alone leads to the induction of poor and erraticimmune responses. This is because Ids are essentially self-antigens, and the immunesystem either ignores them or has been tolerized during prolonged exposure. Oneprinciple we have established is that genetic fusion of Id to a highly imnlunogenicmolecule induces significant anti-Id imlnune responses by providing cognate T cellhelp. OUf initial choice for a fusion partner was the non-toxic FrC of tetanus toxin, aconlpollent of a prophylactic vaccine against tetanus (SpeJlerberg et aI., 1997). ADNA vaccine encoding the Id-FrC fusion antigen induced a dramatic promotion ofanti-Id response, which resulted in protection against tumour challenge in severaltumour models (King et aI., 1998 and unpublished data). This fusion vaccine designis now in clinical trial for patients with a number of B cell malignancies. However,vaccines that include Pre will have to operate in patients with pre-existing immunitydue to vaccinations against tetanus. This could be deleterious, due to clearance of theexpressed antigen. Modelling this situation in mice by pre-vaccination with tetanustoxoid showed that protection can still be induced. However, slowing of the anti-Idantibody responses occurred (Forconi et ai., 2002). This limitation would not apply toantigens to which the immune system has not been exposed either by vaccination orby infection (priInary antigens).

We turned our attention to the group ofcarrier ITIolecules, CPs ofplant viruses. OUf

initial consideration was that it would be unlikely that the human population wouldhave pre-existing imlnunity to these immunogenic molecules, and therefore antigensfused to theln would be presented in the setting of a primary immune response tocarrier molecules. ItnportantlYt plant viral CPs have been used to promote antibodyresponses to linked B cell epitopes in a number of models of infectious diseases(Turpen et ai., 1995; Dalsgaard et al., 1996). The ability of such proteins to self­assemble into viral-like particles in the absence of viral RNA made them theInolecules ofchoice for multiple presentation ofdefined B cell epitopes on the surfaceof large particles (review: Porta and Lomonossoff, 1998). This type of chimericItloIecu)e induces potent immune responses, even without the addition of an adjuvant(McInerney el aI., 1999). The majority of the studies using plant viral CPs as carriermolecules concentrated un inserting short pathogen-derived epitopes t rather thanusing whole antigen sequences. For B cell malignancies, our strategy has been to usethe full length variable fragments of heavy and light chain gene sequences assembledas a single chain (scFv), rather than defined short idiotopes (Hawkins et at., 1994).This induces broader immunity, and eliminates the need for idiotope identification.One particular CP from PYX especially caught OUT attention because a few studies

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 6: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

104 N.SAVELYEVAetal.

indicated that long polypeptides could be fused to the N terminus of this CP whilemaintaining the structural integrity of both fusion partners (Santa Cruz et al., 1996;Smolenska et al. t 1998). As one aim was to identify a fusion partner with no pre­existing antibody in humans~ we tested the sera of normal individuals for antibodiesto PVXCP. We found that all serum samples tested were positive for anti-Freantibodies, but were negative for antibodies to PVXCP (unpublished data). A numberof studies ofprotein vaccination reported that linkage ofB cell epitopes to CP ofplantviruses including PVXCP induced potent antibody responses, preferentially of IgG2aisotype in mice (Brennan et al., 1999; Marusic et ai., 2001). This indicates that thenature of these carrier molecules tends to direct the immune response towards the Th Itype. Therefore, we hypothesized that a DNA fusion vaccine that includes PVXCPwould likely direct the response against attached antigens towards the ThI type. Thus,PVXCP was chosen both to explore the responses induced by DNA fusion vaccinesin the absence of pre-existing immunity, and to determine the effect of an alternatecarrier molecule on the nature of immune response.

Assembly and characterization of Id-fusion vaccines

Individual Id DNA vaccines can be assembled as scFv by linking variable fragmentsofheavy and light chain genes together by peR using family-specific V and J segmentprimers. The fused VH-VL molecule also incorporates a flexible linker sequencebetween the VH and VI. to allow Ids to fold properly (Figure 5.2). The assembled scFvmolecule can be joined to the PVXCP sequence by peRt or by ligating the two genestogether (Figure 5.2). We have also incorporated either a four amino acid (GPGP) orseven amino acid linker (AAAGPGP) between scFv and PVXCP. The latter containsa NotI restriction site to facilitate fusion gene asselnbly. We repeatedly found that theusage of either the four or seven amino acid linker allows the chimeric molecules toretain structural integrity. All our DNA vaccines contain an Ig leader sequence todirect the synthesized protein to the endoplasmic reticulum. Assembled fusion genesare cloned into the pcDNA3 plasmid using HindIII and XbaI restriction sites.Expression is tested by transcription/translation in rabbit reticulocyte lysates andtransfection into Cos- I cells. Cos-l cell supernatants are analysed by ELISA usinganti-PVXCP polyclonal antibody. We also found that chimeric PVXCP fusionproteins can be sedimented by ultracentrifugation. This is due to the ability of scFv­PVXCP fusion proteins to self-aggregate. The pellets can be further analysed byWestern blot analysis using polyclonal anti-PVXCP antiserum, and eJectronnlicroscopy using immunotrapping with anti-PYX IgG. Such an in vitro analysis priorto testing a vaccine in vivo gives a good indication of the size of the chimeric product,levels of secretion, and the state of aggregation. scFv-PVXCP chimeric proteins aregenerally secreted into supernatants at high levels. However, we found that the fusionof small fragments, like a leader sequence to PVXCP, reduced secretion from Cos-Icells when compared to the long scFv fragments attached to PVXCP. For immuniza­tion, DNA vaccines are prepared using a Qiagen giga prep plasmid kit.

Promotion of CD4+ mediated protection against lymphoma and myeloma

The measure of a successful vaccine design against a tumour is the ability to induce

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 7: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include plant virus coat proteins 105

IdiotypicIg

VL

PYX

particle

/ !IIr~~l..l--- C::t~:OpeR amplification~

~

~ peR assembly

L;3o.~~~~.....3r.....::~~V_L-----l 1::::pvx~::::1scFv

Clone intovaccine vector

scFv-PVXCPDNA fusionvaccine

Figure 5.2. Construction of scFv-PVXCP fusion vaccines. Assembled as scFv ~ V nand VI. genes derivedfrom a tumour arc cloned into a DNA vaccine with previously inserted PVXCP gene.

protective responses against tumour challenge. Initially ~ we evaluated the perform­ance of the scFv-PVXCP DNA fusion vaccine design in two murine models of B cellmalignances, the A31 lymphoma and the 5T33 myeloma (Savelyeva et al., 2001). TheA31 lymphoma carries an idiotypic IgM on the cell surface and is MHC class IIpositive. Vaccination of mice with the scFvA31-PVXCP DNA vaccine inducedprotection against challenge with the A31 tumour~ with no effect of either PVXCPalone or scFvA31 alone (Figure 5.30). The protection was accompanied by inductionof low levels of antibody response to idiotypic IgM (typically only half of the miceresponded)~whereas anti-PVXCP antibody responses were high (Figure 5.3b). Anti..Id antibody was of an IgG2a isotype, indicating induction of a Th1 response. Sincepreviously published data demonstrated that antibody was the key effector mecha­nism in protection in a number of Ig-surface positive lymphoma models, the lack ofcorrelation of anti-Id antibody induction and protection was surprising. This sug­gested that other protective mechanisms could be induced in this model. In fact,depletion of CD4+ T cell subsets cOlnpletely abrogated protection, indicating theiInportance of CD4+ T cells in protection against the A31 lymphoma (Figure 5.3c).Passive transfer of the immune sera at the levels induced in vaccinated mice did notconfer long-term protection, though a low level ofprotection was seen, indicating thatthe role for antibody in protection was possible but not adequate (unpublished data).

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 8: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

106 N. SAVELYEVA efll/.

100

80

7ii> 60.~

:1(I) 40"$.

20

p.scFvA31-PVXCP¥

I-a~'l

;.~...

j lL.·l

t_-t-.~=!--··--i: I J L ....., '....·..·1 ~.scFvA31 alone

non· !---1 L. ---- I ~._ _~ _ ~..

Ovaccinated-': ! p.PVXCP alone

~....;;;..,;;;...;;.,,;,a;;';;""';";""-""';':'''''''""--,-:,,_--'''''''''''---rI I • I

20 30 40 50 60 100 110 120

Days after tumour challenge

(a)

(b)C")

(:)

450 90 X

~~ 0 6 ~~~ 350 70 >0-

f 300 60 -g~ ~

~ 250 50 :.ecu 200 .6. 40 :~ A 0~ 150 A 30 ~

; 100 IA 20 tTco ..-c 50 A 10 ~

q,O~-......-~-~~----....--~o

p.scFvA31 p.scFvA31-PVXCP p.PVXCP

o anti..A31ld antibody Aanti-PVXCP antibody

rat IgG¥-~ ;CCinated

-i..------tll !

anti-CD4¥non- __..........---------t t--A

vaccinated.L.-. ....... ~ I----

20 30 40 50 80

Days after tumour challenge

(c)

100

80

~ 60.~

~0 40~III

20

010

Figure 5.3. Performance of the scFv-PVXCP DNA fusion vaccine in the A311ylnphoma nlodel. (a)Induction of protective imtnunity: Juice were vaccinated with 50 IJg of the p.scFvA3I-PVXCP fusionvaccine. or p.scFvA31 alone. or p.PVXCP alone three times at days 0.21 and 42. At day 63. tnice \verechallenged with the A31 (umour, nOll-vaccinated mice \vere included as controls. (b) Antibody responsesto A31 Id and PVXCP at day 35 as ancasured by ELISA. (c) Protection against challenge with the A31tUlnour is dependent on CD4+ T cells. Mice vaccinated with the p.scFvA31-PVXCP fusion vaccinereceived MAbs against CD4+ T cells or rat IgG as a control before and after tUlnour challenge.

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 9: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include !Jlallt virus coat proteins 107

We also demonstrated that the effector mechanism induced by the scFv-PVXCPDNA vaccine against lympholna was different from that induced by the scFv-FrCDNA vaccine where antibody was the key mediator of protection (Save)yeva ef at.,2001). The important lesson learnt from these experiments was that the protectiveeffector lnechanism induced by idiotypic fusion vaccine was influenced by the natureof the carrier nl0lecule fused to the Id.

A similar scFv-PVXCP DNA vaccine design was tested in the 5T33 myelomaInodel that resembles human Inyeloma in that it secretes an idiotypic IgG para­protein, and is surface Ig and MHC class II negative. Since the tumour ceJls, incontrast to lymphoma, do not express Id on the cell surface, we did not expectantibody to playa role in protection in this model. In fact, using Id protein vaccination,we confinned previously that antibodies do not protect against the 5T33 myeJonla.Vaccination with the scFv5T33-PVXCP DNA format, however, induced significantprotection against challenge with the mye]otna (Figure 5.4a). Although the antibodyis not itnportant for protection, it provides a useful indicator of the imlnune response.Generation of antigen-specific IgG in particular is indicative of induction ofantigen­specific T cells, and IgG isotypes are markers of which type of helper cell has beeninduced. The 5T33-PVXCP vaccine induced high levels of anti-5T33 Id and PVXCPantibodies (Figure 5.4b). The antibodies were preferentially of the IgG2a isotype~

consistent with induction of the Th I type of response. Again, as for lymphoma, welooked for cellular effector 11lechanisrns by depletion of CD4+/CD8+ T cell subsets.These results demonstrated that protection in this model was mediated by CD4+ Tcells (Figure 5.4c). Furtherrnore, we have shown that for induction of both antibodyand protective CD4+ T cells, fusion of scFv and PVXCP was required, because aInixture of the two plaslllids encoding each individual component induced neitheranti-Id antibody nor CD4+ T cell-mediated protection.

Mechanism of DNA fusion vaccine operation

Pronlotion of specific anti-Id immunity by vaccination with scFv-PVXCP DNAvaccines is likely to operate through several mechanisms. First, PVXCP, sinli1ar to

FrC, provides cognate T cell help for the idiotype-specific Band T cells. In the caseofB cells, CD4+ T cells specific for PVXCP provide help for idiotype-specific B cellsin a hapten-carrier manner (Figure 5.5). This leads to production of Id-specific IgG inboth models. For induction of Id-specific antibody, activation of PVXCP-specificCD4+ T cells is sufficient, and Id-specific CD4+ T cells are not required. However~these cells are absolutely critical for protection in the A31 lymphoma and the 5T33myelolna nlodels. They appear as a result of T cell help received froln PVXCP­specific CD4+ T cells. Recently, Gerloni and co-workers have demonstrated a novelhelper role for CD4+ T cells, Le. CD4+ T cells specific for a dominant epitope providehelp to CD4+ T cells that recognize an immunologicaHy silenl epitope (Ger)oni et 1I1.~

2000). This is the so-called 'linked T cell help for he)pers~ nlechanism. It involvesaction ofdominant epitope-specific CD4+ cells not directly on CD4+ T cells specificfor the silent detenninant, but through dendritic cells, in a fashion similar to that ofactivation of CD8+ ceIJs with the help of CD4+ T ceHs (Figure 5.5). The help resultsin upregulation ofco...stimulatory molecules on the cognate dendritic cell. The Th-Thco-operation requires that both dOlninant and silent epitopes are presented by the

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 10: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

180 b0t 160

.,...0 )(

140 ~120 ~

~0 100 0

.Q

80~cco

6 60 0..A (,)

0 A 40)(

A >• A A

q.20 ~

c0 <C

E 14:; 12>0­-c.8 10

~ 8co:2 6C")

~ 4'9~ 2c

<C 0 ----_....,IIIMr--------- ~-.........

80

"ii 60>.~

:;,In 40'#.

20

0

25

(b)

7 18Q..... 16x

N. SAVELYEVA etal.(a) 100 --.---rr--'I

:! I••..JI ~ i

I il._.'--~---C"l

L.........H.J:{}l!.l......iii ~- p.scFv5T33 alone! i-:-l' .

non- : t lvaccinate~: !~_... p~PVXCP alone

I I--------t-

35 45 55 65 75 125

Days after tumour challenge

108

p.scFvST33 p.scFv5T33-PVXCP p.PVXCP

o anti-5T33ld antibody 6. anti·PVXCP antibody

(c) anti..CD8

100 ¥vaccinated

80 ¥Ci -1~> 60

~.~ rat IgG:I 40en~Q

20 anti..CD41('.

.,'

0 I~

25 35 45 55 65 75 125

Days after tumour challenge

Figure 5.4. Perfonnance of the scFv-PVXCP DNA fusion vaccine in the 5T33 myeloma modeL (a)Induction protective imnlunity: mice were vaccinated with 50 ,..g of the p.scFv5T33-PVXCP fusiollvaccine, or p.scFv5T33 alone. or p.PVXCP alone three times at days 0,21 and 42. At day 63 mice werechallenged with the 5T33 tunlour, non-vaccinated lnice were included as controls. (b) Antibody responsesto 5T33 Id and PVXCP at day 63 as Ineasured by ELISA. (c) Protection against challenge with lhe 5T33tUlnour is dependent on CD4+ T cells. Mice vaccinated with the p.scFvST33-PVXCP fusion vaccinereceived MAbs against CD4+ or CD8+ T cells or rat IgG as a control before and after tumour challenge.

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 11: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include plant virus coat proteins 109

~ Qi aggregated• scFv-PVXCP'"i fusion protein

~~

MHCU+PVXCp·peptide

~ TCR

help toB-cell

Th-Th helpthrough DC

FigureSitS. Enhancement of iInmunity to Id by fusion to PVXCP. Prirned by DC that have taken up scFv­PVXCP protein.. CD4+ T cells specific for PVXCP derived peptide interact \vith Id-specific B cells thatinternalized the fusion protein through HeR specific for Id. Such interaction together with CD40/CD40Lligation leads to production of antipId antibody. Specific for PVXCP, CD4+ T cells "Iso activate DC byincreasing expression of co-stitnulatory nlolecules. This licenses DC to then interact and activate Id­specific CD4+ T cells. Aggregation of Jd-PVXCP protein enhances recognition by DC.

same APe. This is in agreement with our findings that for induction of protectiveCD4+ T cells, fusion of Id and PVXCP is required. The second nlechanism isattributed to a special feature of the coat protein to self-aggregate. PVXCP-fusedchimeras self-assenlble into multiIneric lTIolecules (Savelyeva et al., 200 I). Suchfusion 1l1olecules closely reselnble the structures fornled by coat proteins ofpotexviruses~ i.e. two layer discs, and stacks of discs. It appears that DC may betterrecognize such nlolecules as they are likely to resemble pathogens. This ITIay act onthe level ofenhanced uptake of the antigen by DC, as PVXCP does not directly changethe Inaturation status of DC (unpubUshed data).

How do CD4+ T cells kill tumours?

The p.scFv-PVXCP vaccine format induces potent protective Id-specific CD4+ Tcell-lnediated responses against the A31 Iympholna and the 5T33 myeloma. ThesetUlnours are not only distinct in the fonn of the Id antigen expressed, being surface­expressed for the lynlphorna and secreted for the nlye)onla.. There is also a differencein terms of imlnune recognition by antitumour Id-specific CD4+ T cells, since the

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 12: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

110 N. SAVELYEVA et al.

DIRECTKILLING

Tumourcell

Natural killers.macrophages,neutrophils,eosinophils

Downstreameffector cells

PerforinJgranzyme

granzyme••

TWEAIt"""-.....--- "",",:~"iiiII.DR3TRAIL-R2

~._. .11_-- (DR4)/TRAIL-R3(DR5)

....t:J••IlFas••

CYtokin~release ~\)IFN-y,IL-4

INDIRECTKILLING

EffectorCD4+Tcell

."igllre 5.6. The role of CD4+ T ceUs in antitumour immunity. CD4+ T cells kill tumours acting directlythrough interaction of ligands; FasL, TRAIL, and TWEAK with death receptors (Fas1 TRAIL-R2IR3..DR3), or by perforinlgranzyme-medialed lysis. C04+ T cells can also act indirec[ly through cytokinerelease (IPN-yand IL-4) and engagementofdoWnSlreaIn effectorcells. such as natural killers, macrophages,neulrophils, and eosinophiJs.

lymphoma is MHC class II positive, whereas the myeloma is negative. The impor­tance of anti-Id CD4+ T cells in the effector phase of the immune responseindicates that they playa direct, rather than helper, role in mediating antitumouriInmunity. We have not fully understood the mechanisms of tumour destructionthat are induced by vaccination with scFv-PVXCP DNA vaccines. However, thereare several lnechanisms that can potentially be used by these CD4+ cells fortumour attack (Figure 5.6). The most common mechanisnl that can be used foreither MHC class II positive or negative tumour is direct killing by Thl CD4+ Tcells via interaction of FasL (CD95L) on activated CD4+ T cells t and Fas (CD95)on the target cell (Ju et aI., 1994). For Fas-negative targets and for targets that areFas-positive but non-responsive to Fas signals, which is often the case for tumours,CD4+ T cells can lyse target cells through two other pathways. These involve othermembers of TNF-receptor ligand family, nalnely TRAIL, the ligand for Apo2(TRAIL-R2 and R3, also known as DR4 and DR5 respectively) (Thomas andHersey, 1998; Dorothee et al.~ 2002)" and TWEAK with Apo3 (DR3) as a receptor(Kaplan et al., 2000). Recent evidence also indicates that CD4+ T cells also useperforinlgranzyme-mediated cytotoxity against MHC class II positive tumours(Dorothee et al., 2002).

Indirect antitumour immunity can be induced by activated CD4+ T cells throughproduction ofcytokines, with IFN-y(Th1cells) and IL-4 (Th2 cells) playing a key rolein engaging the downstream effector cells, such as natural killers, granulocytes, andmacrophages (Hung et at., 1998; review: Pardoll and Topalian, 1998). A broader rolein antitunlour immunity has been implicated for IFN..y. This includes a direct

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 13: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include /Jlant virus coat proteins III

cytotoxic effect on tumour cells, upregulation ofMHC class I and Fas expression, andinhibition of angiogenesis (Spets et aI., 1998; MUlnberg et ai., ]999; Qin andBlankenstein, 2000; Beatty and Paterson t 2001).

Concluding remarks and future prospects

There are several approaches used for activating tumour imJTIunity by DNA vaccinesthat utilize TA. They include fusion or co-delivery of cytokines (Syrengelas et ai.,1996), chenl0kines (Biragyn et a!., 1999) or co-stilTIulatory molecules (Conry et aI.,1996), and sonle other approaches (review: Zhu ef at., 2001). The approach we haveadopted is to fuse our TA of interest, Id, to highly immunogenic sequences derivedfrom either a human pathogen, like Fre, or a plant pathogen, like PVXCP. Theprinciple of this is that CD4+ T cells specific for a pathogenic sequence providecognate help for Id-specific CD4+ Band T cells. The ability of PVXCP to aggregatecontributes further to itnmunogenicity of the scFv-PVXCP vaccine design, and thelack of pre-existing itnmunity is an additional advantage of this design when appliedto patients. An inlportant lesson is that immune responses to tumours can bemanipulated by attaching different immuno-enhancing sequences to TA. Anotherfusion vaccine design recently developed in our laboratory further supports thesefindings. This design utilizes a truncated FrC molecule which activates cytotoxicCD8+ to a defined MHC class I epitope derived from TAs (Rice et aI., 2001, 2002).Furthermore, our findings emphasize a much broader role for CD4+ T cells inantitumour imlnunity than sinlply providing help for B cells and T cells, Le. directinvolvement in tUlnour destruction either by a direct or indirect mechanism. ThescFv-PVXCP fusion design provides the opportunity to activate CD4+ mediatedimmunity, which can act alone or potentially synergistically with vaccines activatingother arms of the inlffiune response.

In the future, incorporation of an alpha viral replicon, an RNA polymerase thatcan alnplify the antigen-encoding RNA, may allow further improvement of per­formance of DNA fusion vaccines that include imlnuno-enhancing sequences fusedto Id (review: Polo et ai., 2000). Another promising approach is a heterologouspritne/boost reginle. In this case1 chimeric PYX viral particles that display Id ontheir surface can be used as a booster vaccine after priming with the scFv-PVXCPDNA vaccine.

To address the problem arising from the technical demand of individual Id vaccineassenlbly, universal antigens, which can be included into a generic fusion 'off theshelf' vaccine against B celllunlours, need to be identified. With the success of DNAInicro-array technology, proteomics and sequencing of the human genome, more ofthis type afTA will be revealed.

Acknowledgements

We thank Dr. Kathy Potter for critical reading of the manuscript. This work wasfunded by the Leukaemia Research Fundt UK.

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 14: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

112 N. SAVELYEVA et all

References

BEA1TY~ G. AND PATERSON~Y. (2001). IFN-gamma-dependent inhibition of tumour angio­genesis by tumour-infiltrating CD4+T cells requires tumourresponsiveness to IFN-gamma.Journal ofb1'll111l1101ogy 166, 2276-2282.

BIRAGYN~A., TANI, K., GRIMM, M.C., WEEKS, S. AND KWAK, L.W. (1999). Genetic fusion ofchemokines to a self tumour antigen induces protective, T cell dependent antitumourimOlunity. Nature Biotechnology 17, 253-258.

BOT, A., STANt A.C., INADA, K. t STEINMAN, R. AND BONA, C. (2000). Dendritic cells at a DNAvaccination site express the encoded influenza nucleoprotein and priIne MHC class 1­restricted cytolytic lymphocytes upon adoptive transfer. brterlltlt;Olltlll11l1llUIlO[ogy 12,825-832.

BRENNAN t F.R., jONE..~, T.D., LONGSTAFF~ M. ET A.L. (1999). Immunogenicity of peptidesderived froln a fibronectin-bindillg protein of S. aurellS expressed on two different plantviruses. Vllccine 17, 1846-1857.

CAMPBELL, MJ., CARROLL, W., KON, S. ET I\L. (1987). Idiotype vaccination against murine Bcell lyrnphoma. HUlnoral and cellular responses elicited by tumour-derived imlnuno­globulin M and its molecular subunits. Journal ofI11l1tlUIl0[Ogy 139, 2825-2833.

CONDON, C., WATKINS. S.C., CELLUZZI, C.M., THOMPSON, K. AND FALO, L.D., JR. (1996).DNA-based hnlTIunization by in vivo transfection of dendritic cells. Nclture Medicine 2,1122-1128.

CONRY, R.M., WIDERA, G., LOBUGLIO, A.F. ET AL. (1996). Selected strategies to auglnentpolynucleotide immunization. Gene Therapy 3, 67-74.

DALSGAARD, K., U·n'ENTHAL, A., JONES, T.D. 1:.7 AL. (1996). Plant-derived vaccine protectstarget anhnals against a viral disease. Nature Biotechnology 15, 248-252.

DOROTHEE, G., VEl~GNONt I., MENEZ, J. ETAL. (2002). Tumour-infiltrating CD4+Tlytnphocytesexpress AP02 ligand (AP02L)lfRAIL upon specific stinlulation with autologous lungcarcinoma cells: role of IFN-alpha on AP02LITRAIL expression and -mediated cytotox­icity. }Ollr'llli of111linUIlO!ogy 169, 809-817.

FELTQUATE, D.M., HEANEY, S., WEBSTER, R.G. AND ROBINSON, H.L. (1997). Different Thelper cell types and antibody isotypes generated by saline and gene gun DNA immuni­zalion. Jourllal OJ'!71l11ZUIl0[Ogy 158, 2278-2284.

FORCONI~ F., K(NG~ C.A., SAHOTA, S.S., KENNA\VAY, C.K., RUSSELL, N.H. AND STEVENSON,F.K. (2002). Insight into the potential for DNA idiotypic fusion vaccines designed forpatients by analysing xenogeneic anti-idiotypic antibody responses. lnl1ll1111o logy 107,39­45.

Fu, T.M., ULMER, J.B., CAULFIELD, MJ. £1'I\L. (1997). PriJning ofcytotox.ic T lymphocytes byDNA vaccines: rcquirclnent for professional antigen-presenting cells and evidence forantigen transfer frollll11yocytes. Molecula!" Medicine 3, 362-371.

GeORGE, AJ., TUTT, A.L. AND STEVENSON, F.K. (1987). Anti-idiotypic inechanisms involvedin suppression ofa Inouse B celllynlphoma. BeL1. Journal ofl11Zl1llI1l0/()1,.S:Y 138~ 628-634.

GERLONI, M., XIONG, S., MUKERJEE, S.~ SCHOENBERGER, S.P., CROFT, M. AND ZANEn~lt M.(2000). Functional cooperation between T helper cell detenninants. Proceedings 0.1 theNatio1lal Acadel11)' ofSciences of/he United States ofA11lel"ica 97, 13269-13274.

HA\VK1NS~R.E., ZHU, D., OVECKA, M. ETAL. (1994). Idiotypic vaccination against hUluan Bcelllylnpholna. Rescue of variable region gene sequences froln biopsy Jnaterial for assemblyas single-chain Fv personal vaccines. Blood 83, 3279-3288.

HEMMI, H., TAKEUCHI, 0., KA\VAI, T. ET/lL. (2000). A Toll-like receptor recognizes bacterialDNA. Nature 408~ 740-745.

IislJ, FJ.,. CASPAR, C.B., CZER\VINSKI, D. £1'1\1... (1997). Tlunour-specitic idiotype vaccines inthe treallnenl ofpatients \vith B cclllyJnpholna -long-tenn results ofa clinical trial. Blood89,3129-3135.

HUNG, K., HAYASHI, R., LAFOND-WALKER, A. £TAL. (1998). The central role ofCD4+ Teellsin the antitumour ilnnlune response. Journal ofExperiJllental Medicine 188, 2357-2368.

JU, S.T., CUI, H., PANKA, DJ., En"INGER, R. AND MARSHAK-ROTHSTEIN, A. (1994). Partici­pation of target P'IS protein in apoptosis pathway induced by CD4+ Th 1 and CD8+

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 15: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

Engineering DNA vaccines that include plant virus coat !Jroleills 113

cytotoxic T cells. Proceedings of the National Acadel1z)' ofSciences ofthe United SteltesofAnJeric1l91~4185-4189.

KAPLAN, MJ., RAY, D., MO, R.R., YUNG~ R.L. AND RICHARDSON, B.C. (2000). TRAIL{Apo2ligand) and TWEAK (Apo3 ligand) mediate CD4+ T cell killing of antigen-presentingmacrophages. Journal ofInlP1Z11l1o[ogy 164, 2897-2904.

KING, C.A., SPELLERBERG, M.B., ZHU, D. ETAL. (1998). DNA vaccines with single chain Fvfused to fraglnent C of tetanus toxin induce protective imlnunity against lymphoma andmyeloma. Nature Medicine 4, 1281-1286.

KRIEG, A.M. (2002). CpO motifs in bacterial DNA and their iJnmune effects. Annual Reviel1'ofb,J11Utllolo8Y 20, 709-760.

KRIEG, A.M., YI, A.K., MATSON, S. ET AL. (1995). CpG mo~ifs in bacterial DNA trigger directB cell activation. Nature 374~ 546-549.

MARUSIC, C.~ RIZZA, P., LATI'ANZI, L. Ef AL. (2001). Chimeric plant virus particles asirnrnunogens for inducing lTIurine and hlunan immune responses against human immuno­deficiency virus type I. Journal ofV;rology 75, 8434-8439.

McINERNEY, T.L., BRENNAN, F.R., JONES, T.D. AND DIMMOCK, N.J. (1999). Analysis of theability of five adjuvants to enhance immmune responses to a chimeric plant virusdisplaying an HIV-1 peptide. Vaccine 17~ 1359-1368.

MU~1BERG,D., MONACH, P.A., WANDERLING~ S. ET AL. (1999). CD4+ T cells eliminate MHCclass II-negative cancer cells in vivo by indirect effecl of IPN-y. Proceedings of tlteNational Acadenzy ofSciences (~f the United States ofA,ller;ca 96, 8633-8638.

PARDQLL, D.M. AND TOPALIAN, S.L. (1998). The role of CD4+ T cell responses in antitumourirnlnunity. Current Opin;01l in Inllllultology 10, 588-594.

POLOT J.M., GARDNER, J.P., JI, Y. l:.T AL. (2000). Alphavirus DNA and particle replicons forvaccines and gene therapy. Developl1Jen1al Biology (Basel) 104, 181-185.

PORTA, C. AND LOMONOSSOFF~G.P. (1998). Scope for using plant viruses to present epitopesfroln animal pathogens. Rev;e~vs ;n Medical Virology 8, 25-41.

QIN, Z. AND BLANKENSTEIN, T. (2000). CD4+T cell-mediated tumour rejection involvesinhibition of angiogenesis that is dependent on IFN-y receptor expression by 110n­

haematopoietic cells. It,111Ufn;ty 12, 677-686.RICE, J., ELLJOlT, T., BUCHAN, S. AND STEVENSON, F. K. (200) ). DNA fusion vaccine designed

to induce cytotoxic T cell responses against defined peptide rnotifs: irnplications for cancervaccines. Journal ofInlnzll11olo8Y 167t 1558-1565.

RIC~ J., BUCHAN, S. AND STEVENSON, F.K. (2002). Critical cOlnponents of a DNA fusionvaccine able to induce protective cytotoxic T cells against a single epitope of a tumourantigen. Journal of ltll11UlIlO!Ogy 169, 3908-3913.

SANTA CRUZ, S., CHAPMAN, S., ROBERTS, A.G., ROBERTS, I.M., PRIOR, D.A.M. AND OPARKA~

KJ. (1996). Assembly and movenlent ofa plant virus carrying a green fluorescent proteinovercoat. Proceeding.s· ofthe NCJliollaLAc(.uJenl)1 a/Sciences o.fth.e UllitedStates ofAl1lericCl93, 6286~6290.

SAVELYEVA, N., MUNDAY, R., SPELLERBERG, M.B., LOMONOSSOFF, G.P. AND STEVENSON,F.K. (2001). Plant viral genes in DNA vaccines activate linked CD4+ T cell mediatedimmunity against B cell JllaHgnancies. Nature Biotechnology 19, 760-764.

SMOLENSKA, L., ROBERTS, LM., LEARl\10NTH, D. £1' AL. (1998). Production of a functionalsingle chain antibody attached to the surface of a plant virus. FEBS Letter.'} 441, 379­382.

SPELLERBERG, M.B., ZHU., D., THOMPSETT, A., KING, C.A., HAMBLIN, T.J. AND STEVENSON,F.K. (1997). DNA vaccines against lymphoma. Prornotion of anti-idiotypic antibodyresponses induced by single chain Fv genes by fusion to tetanus toxin fragment C. Journalofbllmuflo!ogy 159, 1885-1892.

SPETS,. H. t GEORGII-HEMMING, P., SILJASON, J., NILSSON, K. AND JERNBERG-WIKLUND, H.(1998). FaslAPO-l (CD95)-mediated apoptosis is activaled by interferon-gamlna andinterferon- in interleukin-6 (IL-6)-dependent and IL-6-independent ITIu1tipIe rnyelorna cellJines. Blood 92, 2914-2923.

SVRENGELAS, A.D., CHEN, T.T. AND LEVY, R. (1996). DNA imnlunization induces protectiveimlnunity against B celllymphoina. Nature Medicine 2, I038~1041..

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014

Page 16: Engineering DNA Vaccines that Include Plant Virus Coat Proteins

114 N. SAVELYEVA et al.

TANG~ D.C., DEVlT:o M. AND JOHNSTON~ S.A. (1992). Genetic ilnlTIUnizatioll is a siluple luethodfor eliciting an inlmune response. Nature 356, 152~154.

THOMAS, W.D. AND I-iERSEY:o P. (1998). TNF-related apoptosis-inducing ligand (TRAIL)induces apoptosis in Fas ligand-resistant Inelanoma cells and Inediates CD4 Teen killingof target cells. Journal oj'IJ11111111l0/ogy 161, 2195-2200.

TURPEN, T.:H., REINL, 5.1., CHAROENVIT, U., HOFFlvIAN, S.L., FALLARME:o V. AND GRILL, L.K.(1995). Malarial epitopes expressed on the surface of recoillbinant tobacco l110saic virus.Bioffeclrnology 13, 53-57.

ZHU, D., RICE, J., SAVELYEVA, N. AND STEVENSON, F.K. (2001). DNA fusion vaccines againstB cell tUlnOlJrS. Trends in Molecular Medicine 7t 566-572.

Dow

nloa

ded

by [

Uni

vers

ity o

f L

ouis

ville

] at

18:

47 2

0 D

ecem

ber

2014