Accepted Manuscript Bacterial toxin's DNA vaccine serves as a strategy for the treatment of cancer, infectious and autoimmune diseases Elham Behzadi, Raheleh Halabian, Hamideh Mahmoodzadeh Hosseini, Abbas Ali Imani Fooladi PII:
S0882-4010(16)30387-4
DOI:
10.1016/j.micpath.2016.09.017
Reference:
YMPAT 1957
To appear in:
Microbial Pathogenesis
Received Date: 16 July 2016 Revised Date:
18 September 2016
Accepted Date: 21 September 2016
Please cite this article as: Behzadi E, Halabian R, Hosseini HM, Imani Fooladi AA, Bacterial toxin's DNA vaccine serves as a strategy for the treatment of cancer, infectious and autoimmune diseases, Microbial Pathogenesis (2016), doi: 10.1016/j.micpath.2016.09.017. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
1
Bacterial Toxin’s DNA Vaccine Serves as a strategy for the Treatment of Cancer,
2
Infectious and Autoimmune diseases
3 Elham Behzadi1, Raheleh Halabian1*, Hamideh Mahmoodzadeh Hosseini1, Abbas Ali Imani
5
Fooladi1*1
6 7
1.
RI PT
4
SC
Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
8
M AN U
9 10 11 12
TE D
13 14 15
EP
16
AC C
* Corresponding Authors:
Abbas Ali Imani Fooladi, Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran. Email:
[email protected] and
[email protected]
Raheleh Halabian, Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran. Email:
[email protected] and
[email protected]
1
ACCEPTED MANUSCRIPT
17 Absract
19
DNA vaccination –a third generation vaccine- is a modern approach to stimulate humoral and
20
cellular responses against different diseases such as infectious diseases, cancer and
21
autoimmunity. These vaccines are composed of a gene that encodes sequences of a desired
22
protein under control of a proper (eukaryotic or viral) promoter.
23
Immune response following DNA vaccination is influenced by the route and the dose of
24
injection. In addition, antigen presentation following DNA administration has three
25
different mechanisms including antigen presentation by transfected myocytes, transfection
26
of professional antigen presenting cells (APCs) and cross priming.
27
Recently, it has been shown that bacterial toxins and their components can stimulate and
28
enhance immune responses in experimental models. A study demonstrated that DNA
29
fusion vaccine encoding the first domain (DOM) of the Fragment C (FrC) of tetanus
30
neurotoxin (CTN) coupled with tumor antigen sequences is highly immunogenic against
31
colon carcinoma. DNA toxin vaccines against infectious and autoimmune diseases are less
32
studied until now.
33
All in all, this novel approach has shown encouraging results in animal models, but it has to go
34
through adequate clinical trials to ensure its effectiveness in human. However, it has been proven
35
that these vaccines are safe, multifaceted and simple and can be used widely in organisms which
36
may be of advantage to public health in the near future.
37
This paper outlines the mechanism of the action of DNA vaccines and their possible application
38
for targeting infectious diseases, cancer and autoimmunity.
39
Key words: DNA vaccines; infectious diseases; cancer; autoimmunity
AC C
EP
TE D
M AN U
SC
RI PT
18
2
ACCEPTED MANUSCRIPT
40 41 High Light:
43
• DNA Toxin Vaccine and Bacterial Toxin
44
• DNA Toxin Vaccine as a Treatment Option
EP
TE D
M AN U
SC
Contents 1. Introduction 2. Bacterial toxins and immune system responses 3. Mechanism of action of DNA toxin vaccine 4. DNA toxin vaccine for treatment of infectious diseases 5. DNA toxin vaccine for cancer therapy 6. DNA toxin vaccine for autoimmune diseases 7. Concluding remarks and future perspectives 8. Acknowledgement
AC C
45 46 47 48 49 50 51 52 53 54
RI PT
42
3
ACCEPTED MANUSCRIPT
1. Introduction
56
The recent concern of researchers is treating the infectious diseases, cancer and autoimmunity by
57
developing new vaccines that help to elicit stronger immune system responses [1-3]. The novel
58
strategy is to use genetic vaccines composed of DNA or RNA, which have immune stimulating
59
properties with no infection risk. On the other hand, the production of new vaccines must be
60
economical and safe. Great improvements in biotechnology and vaccine production have opened
61
the way to construct "smart vaccines" in the coming future [4]. Innovation of naked plasmid
62
vectors expressing desired gene in the host cells encouraged the scientists to use this approach
63
for curing patients, but naked DNA is poorly immunogenic and it is important to be injected in
64
an efficient way that delivers into the target cells [5]. Investigations have shown that there are
65
some parasitic sequences in vertebrates' genome revealing the occurrence of genomic invasions
66
in their ancestors. This finding shows that human immune system may not recognize naked
67
delivered DNA in vaccine unless it is translated into specific proteins; which must be processed
68
and presented by the professional antigen-presenting cells (APCs) to elicit an antigen-specific
69
immune response [6]. One of the advantages of DNA vaccination is providing sustained
70
exposure of APCs to low levels of antigen to develop appropriate responses [7].
71
A number of research studies in 1990s [8, 9] showed the growing interest for DNA vaccine
72
technology, but the most prominent research made by Wolff et al. proving that direct
73
intramuscular administration of DNA and genetic information led to expression of transgene and
74
stimulated immunological response within the muscle cells [10]. Since then the naked plasmid
75
DNA has been used as a tool to treat many diseases. This approach can induce antibodies as
76
well as cellular responses and can alter cytokine profile. This technology has not the
AC C
EP
TE D
M AN U
SC
RI PT
55
4
ACCEPTED MANUSCRIPT
shortcomings of viral vectors in stimulating unfavorable immune response and the risk of
78
mutagenesis [11].
79
On the other hand, the delivery mode of DNA vaccines determines the level of induced
80
immunization in an individual. Therefore, new generation of DNA vaccines are co-delivered
81
with Micro- or Nano-particles [11]. Recently DNA vaccine encoding certain domain of a
82
bacterial exotoxin has been demonstrated to enable translocation from the extracellular part into
83
the cytoplasm and therefore greatly enhance vaccine potency for curing diseases such as cancer
84
[12], infectious diseases [13] and autoimmune diseases [14]. In some cases the injection of DNA
85
and sequential expression of the gene may induce cytotoxic T lymphocytes (CTLs) to demolish
86
transfected cells [15] or in some cases may cause anti-DNA antibody response and lead to Lupus
87
like syndrome [16].
88
This review focuses on the opportunities offered by DNA toxin vaccines for the treatment of
89
diverse diseases. The applications and the mechanisms of action of some most promising
90
therapeutic DNA toxin vaccines are also described in this review to open the way for designing
91
new toxin-derived molecules for medical treatments.
92
2. Bacterial Toxins and Immune System responses
93
Bacterial toxin is a virulence factor, mostly deleterious in human and other bacterial host
94
produced by both gram positive and gram negative bacteria. These toxins target surface receptors
95
on cells and injure cells or sometimes kill the host [17]. Bacterial toxins can modulate signaling
96
pathways implicated in different physiological events [18]. The host immune system adopts
97
different strategies to combat the effect of toxins and most efficient mode is producing the
98
antitoxin antibodies. These antibodies are of essential importance against the bacteria. Normally,
AC C
EP
TE D
M AN U
SC
RI PT
77
5
ACCEPTED MANUSCRIPT
99
detoxified bacterial toxins are treated with chemical agents or modified by genetic handling to develop various vaccines [19].
101
Bacterial toxins can also elevate the immune responses to other irrelevant antigens specifically
102
when delivered by mucosal routes [20]. Nowadays bacterial toxins and their nontoxic
103
components are used as DNA fusion vaccines and mucosal adjuvant in subunit vaccines against
104
various diseases such as infections [21], autoimmune diseases [22] and cancer[23].
105
Bacterial toxin affects immediately innate or non-specific immune system but can also activate
106
adaptive or specific (antigen-specific T and B cells) immune system [19].
107
Immunogenicity of killed and attenuated bacterial vaccines is partly assigned to adjuvant activity
108
of toxins in association with other cell wall components of bacteria [24].
109
DNA fusion gene vaccines are poorly immunogenic but by the fusion to the bacterial toxin
110
genes, their immunogenicity can be enhanced and stimulate the innate immune system,
111
effectively. In this approach, the main effort is to modify toxin gene in a way that their toxicity
112
diminish but still are able to elicit immune responses [25]. In a study, Pizza et al. produced
113
several Escherichia coli heat-labile enterotoxin (LT) mutants with site directed mutagenesis
114
which were completely non-toxic or with highly reduced toxicity but their adjuvanticity
115
remained very strong at mucosal level [26]. Then Arrington et al. designed two plasmid vectors
116
encoding the A and B subunits of LT for assessing their potential as a genetic adjuvant for fusion
117
DNA vaccine which strongly enhanced T helper 1 (Th1) and T helper 2 (Th2) cytokine
118
responses in animal models [27].
119
The innate immune system recognizes and activates by the interaction of specific molecules on
120
microbial pathogens (pathogen-associated molecular patterns or (PAMPs)) with the
121
recognition receptors on macrophages and dendritic cells (DCs) called pattern recognition
AC C
EP
TE D
M AN U
SC
RI PT
100
6
ACCEPTED MANUSCRIPT
receptors (PRRs). These cells function as antigen-presenting cells (APCs) for T cells [28]. Some
123
microbial pathogen receptors are CD14 receptors, Toll-like receptors (TLRs) [29] (Fig 1.), Fc
124
receptors, complement receptors, mannose binding protein, serum amyloid P and C-reactive
125
proteins [30]. Each microbial product is recognized by a specific receptor; for instance NOD-like
126
receptor (NLR) recognizes bacterial toxins [31] (Fig 1.) and TLR9 recognizes CpG motifs in
127
bacterial plasmid DNA [32].
128
As it is shown in figure 1, PAMPs are recognized by TLRs, and then the synthesis of Pro-
129
Interleukin (IL)-1β is stimulated through nuclear factor kappa-light-chain-enhancer of
130
activated B cells (NF-κB). One of the means that bacteria and bacterial products can go through
131
cell membrane is via pore-forming toxin. Cytosolic PAMPs activates NOD-like receptors
132
(NLRs) which cause formation of caspase-1-activating inflammasomes. The inflammasome
133
adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) is
134
recruited to activate caspase-1. The role of caspase-1 is to induce the cell necrosis or pyroptosis
135
and usually interfered with various developmental stages [31].
136
When bacterial toxin binds to NLR, signals convert through an adaptor protein and some
137
immune response genes and co-stimulatory molecules are transcribed by transcription factors.
138
When PRRs like toxins ligate on APCs, the intensity of antigen presentation in these cells
139
enhance and elicit the T cell-mediated responses. As the protective immunity depends on
140
induction of T helper (Th1 or Th2) responses, the Th2 cells mediate immunity against
141
extracellular bacteria and function as helper for the production of neutralizing antibodies such as
142
IgG, IgE and IgA against bacterial toxins. Macrophages and DCs secrete IL-12, IL-18, IL-23 and
143
IL-27 in response to some microbial products [33].
AC C
EP
TE D
M AN U
SC
RI PT
122
7
ACCEPTED MANUSCRIPT
When DCs are activated by bacterial products especially gram negative bacterial
145
lipopolysaccharides (LPS) the differentiation of T helper cells from immature precursors occur
146
[34]. In response to some bacterial toxins, DCs may release IL-10 and IL-12 production is
147
hindered which subsequently induce IL-10-secreting regulatory T cells (Tregs). Tregs are a kind
148
of T cells with regulatory and suppressive function that control the Th cells and inflammatory
149
responses through attacking the foreign antigen. In this case, the signal for maturation of the
150
DCs may be supplied by interaction between bacterial toxins and PRR and activation of
151
signaling pathways launch the transcription of genes involved in the immune responses. The
152
maturation of DCs and activation of signaling pathways are essential for the onset of T cell
153
responses [35]. Immature DCs in contact with foreign antigens in tissues produce chemokine
154
receptors CCR5, CCR6, CD80, CD86 and class II major histocompatibility complex (MHC) and
155
in this level these cells catch the foreign antigens and engulf them which cause to
156
downregulation of CCR5 and CCR6 expression and CCR7 upregulation. In this stage DCs are
157
placed in the lymph nodes and become mature [36]. CpG motifs which are prevalent in bacterial
158
DNA or LPS may serve as maturation signal for these cells [37]. As DCs mature, the uptake and
159
process the antigen by the DCs diminish but the expression of CD80, CD86 and class II MHC
160
molecules elevate the antigen presentation potency. For performing as APCs for Th1 and Th2,
161
DCs must be influenced by the activation signals from pathogens [6]. Some toxins like cholera
162
toxin (CT) produced by Vibrio cholera composed of an enzymatic part (A subunit) and a
163
receptor binding part (five copies of the B subunit) can activate DCs which in turn induce
164
Th2 cells while LPS initiate maturation of DCs that activate Th1. Some bacterial products which
165
work as immunomodulatory molecules can elicit acquired immune responses. These molecules
166
can control cytokine production and maturation of DCs by directing T cell differentiation [38].
AC C
EP
TE D
M AN U
SC
RI PT
144
8
ACCEPTED MANUSCRIPT
Interaction of the antigen-specific T cell receptor (TCR) with peptide-MHC and co-
168
stimulation of CD28 molecules on the T cells and CD80 or CD86 on APCs activate T cells
169
of the acquired immune system [39]. As TCRs have not enough intracellular domains, they
170
associate with CD3 molecules and form TCR/CD3 complex. This complex activates
171
transcription of cytokine related genes such as IL-2. For the production of IL-10, inducible co-
172
stimulator (ICOS) that is expressed on activated T cells binds to B7-H2 on APCs. Other
173
interactions between activated T cells and other co-stimulatory molecules on APCs are possible
174
to form immune synapse. Some bacterial derived molecules can elevate or demolish the co-
175
stimulation of T cells by APCs. Therefore for increasing immune system responses, detoxified
176
bacterial components such as bacterial toxin are employed as adjuvants in biotechnology and
177
vaccine development. Bacterial toxins bind to the T cells and activate them in a polyclonal
178
fashion. Studies show that AB toxins like Pertussis Toxin (PT) and CT can interact with T cells
179
and modify cytokine secretion [40].
180
Moreover, CTLs that are important functional immune effectors which recognize bacterial
181
components on the surface of contaminated cells play significant role in elimination of bacteria.
182
In contrast to CD4+ T cells that identify exogenous antigen in cooperation with class II MHC
183
molecules, CTL identify endogenous antigen in cooperation with class I MHC molecules. To
184
elicit CTL response, exogenous antigens have to go through an endogenous processing pathway
185
to be presented to class I-restricted T cells. Studies have shown that bacterial toxins fused with
186
foreign antigens can activate class I processing pathway [40, 41].
187
As mucosal tissues are the main routes of pathogen entry into the host, nasal or oral delivery of
188
vaccines and induction of local immunity at these sites is the best way to prevent some infectious
189
diseases. Mucosal vaccination improves local immune responses and the effect of parenteral
AC C
EP
TE D
M AN U
SC
RI PT
167
9
ACCEPTED MANUSCRIPT
immunization. Mucosal defense is including the epithelium, mucus, innate and adaptive immune
191
responses. Secretory IgA is important for defending against invasive pathogens and serum IgG is
192
necessary for the prevention of systemic infection [42]. When an antigen carries across the
193
mucosal surface into the mucosal lymphoid tissue, the IgA is secreted. When the antigen is
194
processed, the IgA producing cells will locate in mucosal effector sites and will hinder entry,
195
attachment, and replication of pathogens or neutralizing microbial toxins in the next encounters.
196
As mucosal surface is in the constant exposure to the foreign antigens, it becomes unresponsive
197
to prevent unnecessary reactions to benign antigens [43].
198
For producing mucosal vaccines the antigen delivery system must keep antigen from enzymes
199
and improve the acquisition by Mauthner cell (M-cell) and epithelial cells. Addition of an
200
adjuvant such as bacterial toxin elicits innate and adaptive immune responses against antigens.
201
As the most of bacterial toxins are toxic for the human body, they must be detoxified but keep
202
their immunogenic properties [43].
203
3. Mechanism of action of the DNA toxin vaccine
204
One strategy to deliver antigens into cells of individuals is by using viral and bacterial plasmid
205
vectors. They function as a shuttle system to deliver and express desired antigen for stimulating
206
cellular and humoral responses. Genetic vaccines consist of DNA vaccine, RNA vaccine and
207
viral vaccine [44]. Without considering the mode of action of a DNA vaccine, it can be
208
employed against various tumor cells, or bacterial infectious diseases [45] or autoimmune
209
diseases [46]. In this paper we just review the properties of DNA vaccine.
210
Nowadays, new approaches such as DNA vaccines are available for more specific treatment of
211
diseases [47]. To clinically use these vaccines, the mechanism of action of the naked DNA
212
vaccines and the adjuvant coupled with DNA must be understood. If the desired protein is not
AC C
EP
TE D
M AN U
SC
RI PT
190
10
ACCEPTED MANUSCRIPT
immunogenic, DNA vaccine encoding this antigen may not produce ideal immune responses.
214
But DNA vaccination is a proper approach to manipulate the antigen sequences to improve the
215
immunogenic property of the desired antigen [48].
216
Generally, DNA vaccines transfect DCs, muscle cells and keratinocytes and activate the innate
217
immune system with the help of PAMPs such as unmethylated CpG-containing sequences.
218
Recently, researchers consider improving four characteristics of DNA vaccines to enhance the
219
immunogenicity including improving stability and potency of transfection, eliciting proper
220
immune response and augmenting the antigen presentation by DCs [49].
221
Mammalian expression plasmids consist of a viral promoter (cytomegalovirus (CMV)
222
immediate-early promoter) to manage transcription and translation in vivo, an adjacent intron A
223
sequence to improve transcription efficacy and stability of mRNA then enhance protein
224
expression, a transcription terminator like bovine growth hormone or rabbit beta-globin
225
sequence, which contain strong polyadenylation signal and usually an antibiotic resistance gene
226
for bacterial selection. For expressing more than one immunogen, multi cistronic vector is
227
constructed. The best immune responses with DNA vaccine is achieved if a very active
228
expression vector is utilized [48].
229
The bacterial DNA performs as PAMPs which react with TLRs and other immunity related
230
molecules to activate the immune cells. As plasmid has the ability to express a desired
231
immunogenic protein, amelioration of the vector design for high protein expression is necessary
232
[49]. One approach to increase protein expression is to make the codon usage of bacterial mRNA
233
as optimal as possible for eukaryotic cells. For optimizing the expression the best method is to
234
change the gene sequence of immunogenic protein and design a more common codon which is
235
used in target cell. The most important concern to induce protein expression is the type of
AC C
EP
TE D
M AN U
SC
RI PT
213
11
ACCEPTED MANUSCRIPT
promoter. Previously the Simian virus 40 (SV40) promoter was usually used but recently it has
237
been shown that the Rous Sarcoma Virus (RSV) has a much better expression rate and CMV
238
function even better than those two mentioned before. For improving expression rate some
239
modifications such as insertion of enhancer sequences, modifications of interons, transcriptional
240
termination sequences and polyadenylation signal are required [50].
241
This kind of treatment has some advantages such as multiple genes incorporation into a vector to
242
ensure the modulation of intracellular ways and modification of antigens. By adding a leader
243
sequence, the antigen can target endoplasmic reticulum (ER) and induce humoral immunity. The
244
reverse transfer of antigen from ER to cytosol and delivery of DNA to APC may increase the
245
possibility to produce CD8+ T cell responses. Some studies show that constructing a recombinant
246
vaccine by fusing single-chain Fv (scFv) of Idiotypic immunoglobulin (Id) to tetanus
247
neurotoxin (CTN) fragment C can stimulate high level production of anti-idiotopic antibodies
248
and give immune protection against lymphoma. CTN is composed of a heavy (B chain) and a
249
light chain (A chain) linked through a disulfide bridge. The digestion of toxin by papain
250
yields two fragments; one of them is fragment C. The non-toxic carboxy-terminal fragment
251
of CTN heavy chain (fragment C) has been used as a valuable tool is construction of DNA
252
vaccine [51, 52].
253
Classical adjuvants such as alum commonly used in vaccines to induce high Th2 response with
254
a small amount or no Th1 response. For improving natural ligands and synthetic agonists for
255
PRRs such as TLRs agonists are novel adjuvants which elicit pro-inflammatory cytokines,
256
chemokines, and type I Interferons (IFNs) which enhance the ability of the host immune
257
system to defeat pathogens [53]. Thus for improving the host immunological responses, DNA
258
vaccines can be fused with immunostimulatory molecules such as TLR agonists [54]. In a study
AC C
EP
TE D
M AN U
SC
RI PT
236
12
ACCEPTED MANUSCRIPT
TLR7/8 agonists were used as adjuvants in combination with cancer vaccines that greatly
260
postponed the occurrence of spontaneous mammary tumors and reduced their incidence
261
compared to the DNA vaccine alone [53].
262
It has been proposed that DNA vaccines provide immunity by three different mechanisms: (1)
263
Somatic cells such as myocytes present DNA-encoded antigens through MHC class I pathway to
264
CD8+ T cells, (2) Professional antigen presenting cells (APCs) such as DCs are transfected by
265
DNA and (3) Transfected somatic cells phagocytized by professional APCs present antigens to T
266
cells to cross-prime immune responses. As myocytes cannot effectively present antigens through
267
MHC class I, therefore the two other mechanisms seem to be of more importance in DNA
268
vaccination. Here, antigen presentation is mediated by MHC class I and MHC class II pathways
269
which induce both cellular and humoral immune responses against antigen. Besides the antigen,
270
unmethylated CpG motifs which present in the plasmid backbone of DNA vaccine are
271
recognized by TLR9 and can provoke DCs to promote efficient cell mediated immune response
272
[3].
273
The route of administration is also of importance in the efficacy of DNA vaccines. Either
274
intramuscular (i.m.) needle immunization or intradermal (i.d.) injection using “gene gun” may be
275
used. Gene gun is a needle free apparatus which is used to inject gold beads coated with DNA
276
vaccine plasmids into the epidermal layer of skin. Studies show that the development of immune
277
response and the extent of transfected cell types are influenced by the mean of injection [55].
278
For summarizing this issue, most common advantages and disadvantages of DNA vaccine and its
279
immunological properties in comparison with traditional vaccines are mentioned in the table 1
280
and Fig 2.
AC C
EP
TE D
M AN U
SC
RI PT
259
13
ACCEPTED MANUSCRIPT
Although DNA vaccination has numerous advantages, some issues have been raised with regard
282
to this approach. Occasionally the integration of vaccine DNA into the genome of the host cell is
283
seen, which may cause mutagenesis and activation of oncogenes. Also the repeated DNA
284
vaccination can prompt autoimmunity. On the other hand, further researches have shown that the
285
possibility for integration is very lower than naturally occurring mutations and increase in anti-
286
DNA antibodies and changes in clinical markers for autoimmunity were not detected [13].
287
As shown in table 1., the DNA vaccine provides an attractive approach over other vaccines with
288
several advantages. Although the major problem to develop favorable DNA vaccines is its low
289
efficacy to induce the immune responses, but its production is very cost-effective, has longer life
290
time, has no risk of infection associated with live-attenuated vaccines or pathogens, well
291
tolerated and has an excellent safety record. Therefore, DNA vaccines are a promising modern
292
approach to protect humans and animals from various diseases.
293
To overcome the low efficacy of DNA vaccines, vector and antigen design, delivery methods
294
have been improved and incorporation of adjuvants and prime boosting strategy have
295
significantly enhanced the immunogenicity of these vaccines.
296
4. DNA toxin vaccine for treatment of infectious diseases
297
Vaccination is one of the best means to overcome infectious diseases, which is named after
298
Vaccinia virus. In 1798, Edward Jenner attempted to prevent smallpox after the injection of
299
obtained cowpox pus to healthy people [56].
300
The first bacterial vaccine was discovered by Louis Pasteur in 1880 that inoculated an old culture
301
of Pasteurella multicida (formerly known as Pasteurella septica) to the chickens and protected
302
them against the aforementioned bacteria [57].
AC C
EP
TE D
M AN U
SC
RI PT
281
14
ACCEPTED MANUSCRIPT
Conventional vaccines in use are usually consist of killed, inactivated or live, attenuated
304
microorganisms, some microbial metabolisms such as detoxified toxins, purified antigens or
305
conjugated polysaccharides. Although these vaccines have eradicated many infectious diseases
306
such as smallpox in many countries [58] but they still have some limitations in handling the
307
virulent organisms. In this context, new approaches are under way to develop subunit and DNA
308
vaccines for smallpox typically composed of vaccinia virus (VACV or VV) membrane proteins
309
that stimulate neutralizing antibodies against the intracellular mature virus and extracellular
310
enveloped virus forms [58].
311
The purified microbial components vaccine which introduced in 1920s was used against
312
diphtheria and tetanus and the toxin of both microorganisms was modified by chemical agents to
313
obtain non-toxic toxoid. These early diphtheria and tetanus toxoids should be considered as the
314
first subunit vaccines. Filtration of broth cultures of Corynebacterium diphtheria (C. diphtheria)
315
and Clostridium tetani (C. tetani) resulted in sterile filtrates substance that were toxic for animals
316
and caused diseases in both animals and human. Then the specific antitoxins produced in horses
317
for passive protection and treatment and toxin/antitoxin mixture was used to produce active
318
protection. Sometimes accidental errors in the preparation of mixture caused terrible outcomes
319
such as contamination of mixture with Staphylococcus aureus caused several deaths. Finally, it
320
was found that formalin can detoxify diphtheria toxin (DT) and CTN yet preserving its
321
antigenicity stimulate neutralizing antitoxin [59]. Early was shown that proteins presented in a
322
solution were less immunogenic than protein particles. That’s why the soluble toxoids of
323
diphtheria and tetanus were absorbed to an alum gel to perform as an adjuvant. Many bacterial
324
components such as LPS, toxin and killed bacteria have been used in animal models but because
325
of safety issues, they have not been approved for use in human beings [60]. Recently, for
AC C
EP
TE D
M AN U
SC
RI PT
303
15
ACCEPTED MANUSCRIPT
improving the quality of this toxin considering safety, efficacy and stability researchers have
327
made some effort for genetic inactivation of diphtheria toxoid [61]. C. diphtheria strain C7-
328
(β197) has a non-toxic mutant form of DT called cross-reacting material (CRM197) which is a
329
potent vaccine candidate. DT is a single polypeptide chain A-B toxin consists of two subunits
330
coupled with disulfide bonds; the B subunit is binding fragment and A subunit is the active
331
site of the toxin. When in the active site a glycine is substituted with a glutamic acid, its
332
toxicity abolishes. Thereby, CRM197 is promising option to diphtheria toxoid [62].
333
For enhancing the immunogenicity of some vaccines bacterial polysaccharides were conjugated
334
to a protein antigen to stimulate T cells. The first attempt for producing conjugated vaccines was
335
made on H. influenza type b and then on pneumococci which showed great success [63]. A
336
recent study on DNA vaccine expressing Pneumococcal surface protein A (PspA) has shown
337
proper protection of mice against an intranasal pneumococcal infection [64]. PspA4 is a protein
338
composed of leader sequence, α-helical region, clade-defining region (CDR), proline-rich
339
region (P – block of proline repeats and N – block without proline) and choline-binding
340
region. This protein is a promising candidate against pneumococcal infection. pspA4A and
341
pspA4Pro were two fragments of PspA4 which were used in this study. pAE vector and
342
DNA vaccine vector pSec-Tag2A were used for expression in E. coli. Mice were immunized
343
with PspA as protein adjuvant with alum or DNA vaccine and IgG2a bound to bacteria for DNA,
344
but not protein immunization. Both protein and DNA vaccines were protected the mice against
345
intranasal lethal bacteria. Antibodies induced by protein and DNA immunizations were distinct
346
both in Fc and Fab.
347
As some microbes cannot be grown easily in vitro, they cause a problem in vaccine development
348
with conventional methods. Then the best procedure to prepare vaccines against these microbes
AC C
EP
TE D
M AN U
SC
RI PT
326
16
ACCEPTED MANUSCRIPT
was recombinant technology. In this approach desired genes are transferred to the genome of an
350
easily grown microorganism such as Escherichia coli, Bacillus subtilis or yeast to express a
351
recombinant product. The most used recombinant product is a surface protein of HBV produced
352
in yeast cells that purified and formed as viral particles which stimulate antibodies to protect
353
against infection. The advantages of these vaccines are ease of growing of the hosts, the
354
possibility to upregulate the production and ease of purification [65].
355
Today modern vaccine approaches have substituted traditional methods and we still expect much
356
more advances in this area. The first attempt to demonstrate the effectiveness of a recombinant
357
DNA plasmid in animals was made during the 90's. The first attempt regarding infectious
358
diseases was made by two separate research teams by Fynan et al. and Ulmer et al. in 1993. They
359
found that immunization of mice with a plasmid DNA could protect them against influenza virus
360
[66].
361
Indeed, the appearance of antibiotic resistant bacteria emerged the need of new generations of
362
vaccines. Although some bacterial vaccines have been developed and are widely used for
363
prevention and treatment in humans; the mortality and morbidity rate among ill human being
364
populations still is high. Therefore, an urgent need is seen for new vaccine approaches [67].
365
Since then, diverse studies have shown that DNA vaccines have many advantages over
366
conventional types of vaccines (Table 1.) but they are barely used in infectious diseases. The
367
results of experiments show that bacterial DNA vaccines are going to become reality in the
368
coming future [13].
369
For the elimination of the endogenous bacteria, the immune responses are commonly cellular and
370
against exogenous bacteria antibodies are produced which are necessary for controlling almost
371
any bacterial infection. The antibody usually produced against bacterial toxins and capsule and
AC C
EP
TE D
M AN U
SC
RI PT
349
17
ACCEPTED MANUSCRIPT
structural proteins. For developing an effective DNA vaccine, the bacterial antigenic epitopes
373
must be determined. For designing a proper structure for the vaccine against bacterial infections
374
the relevant antigens must be selected [13]. The schematic immunological mechanism of DNA
375
vaccine in respiratory infections is demonstrated in Fig 3.
376
The occurrence of multidrug resistant mycobacteria, low efficiency of bacillus calmette-guerin
377
(BCG) vaccine and co-infection with HIV-1, a novel DNA vaccine incorporating HIV-1 p24
378
protein is designed, which provides protection to Mycobacterium tuberculosis (M. tuberculosis)
379
and concurrently stimulate humoral and cellular response against HIV-1 in BALB/c mice. In this
380
experiment four undefined epitopes belonging to T cell including Ag85A, Ag85B, MPT64, and
381
TB10 were investigated and 4 antigens of M. tuberculosis were designated [68].
382
Since constructed bacterial DNA must use the eukaryotic cells' machinery and have to be
383
expressed in a host cell, some problems may occur. These obstacles may arise during the
384
folding, translocation and post translational modifications and cause some undesired
385
results. The main and the most important problem concerning these kinds of vaccines are
386
autoimmune responses following vaccination with capsular polysaccharides of some
387
bacteria which have similar structure to the host cellular carbohydrates. To solve this
388
problem, scientists have developed peptides that can mimic polysaccharide antigens and can
389
elicit T cell-dependent polysaccharide immune responses. But these peptides have some
390
limitations; for instance they are not a good immunogenic molecule and have a low stability.
391
Furthermore, small peptides can break down and become ineffective [45].
392
Also it has been demonstrated that during infection or after vaccination, efficient immune
393
responses are produced against bacterial toxins and these immune responses are usually able to
394
protect individuals against toxin-generating bacteria. Scott et al. have shown that plasmids
AC C
EP
TE D
M AN U
SC
RI PT
372
18
ACCEPTED MANUSCRIPT
encoding the receptor binding domains (RBDs) from Clostridium difficile (C. difficile) toxin A
396
(TcdA) and toxin B (TcdB) were able to generate both anti-RBD antibodies and RBD-specific
397
antibody-secreting cells (Table 2) TcdA and TcdB both have enzymatic, translocation, and
398
receptor-binding domains and each domain has a specific role in cell intoxication [69].
399
In summary, naked DNA has the ability to elicit the immune system against an expressed
400
antigen in infectious diseases, and induced immunity has ability to give protection against
401
infection and the foreign antigens. But there are only a limited number of vaccines available for
402
some bacteria and mostly cannot give complete protection [13]. Moreover, some bacteria toxins
403
such as CT B subunit is able to be used as an adjuvant in combination of another bacterial
404
vaccine toxin [70].
M AN U
SC
RI PT
395
405
5. DNA toxin vaccine for cancer therapy
407
Although conventional vaccines are usually administrated to healthy people to prevent different
408
types of diseases, cancer vaccines are used as therapeutic agents to treat cancer patients and
409
fortify their immune system to combat the disease [71].
410
The first attempt for improving immune system in cancer patients is made by Dr. William Coley
411
in 1891. He injected Streptococcus pyogenes and Serratia marcescens into the cancer tumors.
412
Until now, some bacteria such as BCG which is similar to Coley’s toxin are used to treat cancer
413
patients [72].
414
At the present time, efforts for developing an efficient, safe and long-lasting therapeutic cancer
415
vaccine are ongoing. Cancer vaccines are classified into different groups on the basis of their
416
compositions including tumor cell vaccines, protein/peptide-based cancer vaccines, DC vaccines,
417
and genetic vaccines [73]. Peptide plus adjuvant vaccine has the noncomplex cancer vaccine
AC C
EP
TE D
406
19
ACCEPTED MANUSCRIPT
formulation which is composed of peptide injected intradermal with an immunogenic adjuvant.
419
To produce this kind of vaccine the relevant tumor antigens and their related MHC Class I and
420
MHC Class II epitopes must be determined.
421
Plasmid DNA immunization is another simple strategy for developing cancer vaccine. Similar to
422
peptide vaccines, related target antigens must be detected to constitute the plasmid vector to
423
carry the proper gene sequences. Contrary to peptide vaccines the related epitopes are not
424
required to be defined. Another strategy is recombinant viral vectors which are used to deliver
425
determined tumor antigens through gene transfer. The most important recombinant viral vectors
426
are replication-defective adenoviruses (rAd) and adeno-associated viruses (AAV). The
427
immunogenic ability of these vectors is high. Using recombinant bacteria to deliver determined
428
tumor antigens for gene transfer is another strategy in cancer vaccine production. Salmonella
429
typhimurium and Listeria monocytogenes are commonly used as antigen delivery platforms in
430
this strategy. DCs are professional APCs, and strongly stimulate the naïve T cells. MHC class I
431
and MHC class II molecules are highly expressed by activated DCs to excite CD8+ and CD4+ T
432
cells and also provide co-stimulatory signals for activating T cells. So, DCs represent a very
433
attractive vaccine option for cancer vaccines [74].
434
Conventional vaccines are prepared by killed or attenuated microorganisms, antigens, peptides
435
and tumor cell extracts, but most recently used methods for cancer treatment are based on genetic
436
approaches [75] which are discussed in this paper.
437
In a cancer therapy strategy, autologous tumor antigens are used but they are not capable to
438
induce efficient T helper responses, and CD8+ effector T cells are useless in this context. To
439
conquer this deficiency, Rice et al. co-operated sequences derived from CTN in the same vector
440
as tumor antigen to augment tumor related immune responses [76].
AC C
EP
TE D
M AN U
SC
RI PT
418
20
ACCEPTED MANUSCRIPT
Rice et al. in 2002 have described a DNA fusion vaccine encoding the first domain (DOM) of
442
the Fragment C (FrC) of CTN fused to specific MHC class I-binding epitope sequences in the
443
C-terminus. This structure stimulates high levels of CD41 T cell and the insertion of the
444
tumor-derived epitope help to prime epitope-specific CTLs. In addition, epitope-specific
445
CD81 T cells protect individual from tumor challenge (Fig 4.) [77].
446
Most recently, Stevenson et al. have made the similar trial on DNA vaccines using tumor-derived
447
peptide sequences fused to an engineered domain of CTN was able to suppress tumor growth in
448
prostate cancer patients (Table 3) [78].
449
As numerous of cancer tumors are resistant to conventional anticancer therapies, novel
450
approaches have been developed to overcome the problem. In addition, these new strategies must
451
selectively act on cancer cell and have minimal toxicity for normal cells. The application of
452
bacterial toxins is proved to be an effective strategy for cancer treatment [76, 78].
453
Bacterial toxins are able to destroy cells or modify some activities such as cellular divisions,
454
apoptosis and cellular differentiations. These modifications are related to carcinogenesis and may
455
cause cellular abnormality or corrupt the function of normal cells. Cytolethal distending toxins
456
(CDTs) which consider as cell-cycle inhibitors are found in gram negative bacteria such as
457
Campylobacter jejuni and Salmonella typhi and cycle inhibitor factors (Cif) which cause the
458
inhibition of mitosis are found in enteropathogenic (EPEC) and enterohaemorrhagic (EHEC) E.
459
coli. It has been suggested that aforementioned toxins can interfere with clonal expansion of
460
lymphocytes to balance the immune system. On the other hand, cytotoxic necrotizing factor
461
(CNF) induces DNA replication and proliferation of cells but prevent cell differentiation. Some
462
bacteria such as E. coli can release CNF. When CNF influence, the number of tumor cells
AC C
EP
TE D
M AN U
SC
RI PT
441
21
ACCEPTED MANUSCRIPT
does not increase, instead poly-nuclear cells occur. This event appears because of the ability
464
of toxin to prevent cell differentiation and apoptosis [79].
465
One advantage of bacterial toxins is its less side-effect in comparison with other anti-cancer
466
therapies therefore toxin encoding genes can be applied in DNA vaccine technology to augment
467
the efficacy of this approach [80].
468
Some bacterial exotoxins such as Pseudomonas exotoxin A [81], DT [82] and Clostridium
469
perfringens (C. perfringens) enterotoxin [83] have the ability to bind to the surface antigens of
470
tumor cells and be used as anti-cancer vaccines.
471
DT binds to the heparin-binding epidermal growth factor-like growth factor (HB-EGF) precursor
472
on the surface of cells. Internalization of DT-HB-EGF complex through endocytosis via clathrin-
473
coated vesicles occurs. Following DT post-translational process, catalytically active toxin
474
produced. This toxin ribosylates elongation factor-2 (EF-2) and inhibits protein synthesis. This
475
event leads to cell lysis or apoptosis DT post-translational process catalytically active toxin is
476
produced. The action of the toxin is to ribosylate elongation factor-2 (EF-2). Pseudomonas
477
exotoxin also can ribosylate EF-2 and inhibits the protein synthesis [84]. Denis-Mize et al. have
478
shown that vaccination with DNA encoding Pseudomonas aeruginosa (P. aeruginosa) exotoxin
479
A elicit specific immunity and protected mice from the lethal doses of exotoxin A producing
480
from Pseudomonas [85].
481
C. tetani cleaves VAMP-2 and thereby prevents neurotransmitter to be released from pre-
482
synaptic nerve endings in central nervous system (CNS). Above mentioned bacterial toxins with
483
anti-cancer effects can be employed as therapeutic agents in DNA vaccine strategies and be
484
produced within target cells [86]. Chudley et al. have demonstrated that a fusion DNA
485
vaccine which encoded a DOM of fragment C of CTN which was linked to an HLA-A2-
AC C
EP
TE D
M AN U
SC
RI PT
463
22
ACCEPTED MANUSCRIPT
binding epitopes from prostate-specific membrane antigen (PSMA) was safe and well
487
tolerated. The findings and clinical effects were promising for coming research [52].
488
The main goal of cancer vaccine research is obtaining an effective and long-lasting CTL
489
response. CD4+ T cell is produced through a MHC-dependent pathway which is essential for
490
reproduction of CD8+ T cell responses and sustaining memory throughout DNA vaccination. In
491
the previous study the vaccine produced DOM-specific-CD4+and PSMA27-specific CD8+ T cells
492
which were quite significant.
493
Tang et al. found that administration of plasmids containing human growth hormone (hGH)
494
gene was able to provoke humoral immunity against hGH proposing that pDNA can elicit
495
specific immune responses [66].
496
The results of these findings are encouraging which strongly imply that DNA immunization can
497
be an efficient approach to overcome many types of cancers. But, before becoming widely used,
498
it must still undergo further experiments, such as different second generation DNA vaccine
499
optimization strategies.
500
6. DNA toxin vaccine for autoimmune diseases
501
Autoimmune diseases are conditions that the antigen-specific and organ-specific immune invade
502
on their own and autoreactive immune cells are achievable from these patients. It is supposed
503
that the action of specific T cells and/or antibodies is the cause of this disease, therefore
504
inhibiting the production of these cells and antibodies are of therapeutic value. If the
505
autoimmune disease is caused by the action of specific T cells associated with class II antigens,
506
on the antigen-presenting cell, immunomodulators can compete with the self-antigen causing
507
autoimmune disease and if it is at the level of the T cell receptor, an immune response to the
508
variable α and β chains of these receptors can eliminate the autoimmune reaction[87].
AC C
EP
TE D
M AN U
SC
RI PT
486
23
ACCEPTED MANUSCRIPT
An infection can also induce or launch an autoimmune disease. Studies show that most
510
pathogens produce Tregs that sustain tolerance and prevent autoimmune disease. One of the
511
most studied autoimmune diseases is multiple sclerosis (MS). Multiple sclerosis is a progressive
512
autoimmune disorder that damages the myelin sheath of motor neurons in central nervous
513
system. The injury arises as a result of the inflammation and may cause loss of nerve signal
514
conduction and develop secondary disordered repair or gliosis. Four types of MS are
515
recognized until now: Relapsing-Remitting MS (RRMS), Secondary-Progressive MS (SPMS),
516
Primary-Progressive
517
manifestations in all four types are different which indicate localization, the nature and mode of
518
activation of invading cells and recovery process of diseases [88].
519
Smith et al. compared M. tuberculosis that induces a prototypical Th1-mediated immune
520
response with Citrobacter rodentium (C. rodentium), a bacterium that requires IL-23-dependent
521
Th17 cell response during infection to produce experimental autoimmune encephalomyelitis.
522
Mice immunized with C. rodentium adjuvant showed classical signs of experimental
523
autoimmune encephalomyelitis (EAE), similar to the mice immunized with M. tuberculosis, but
524
the disease was not as severe as the latter with moderate progression and the number of Th1 and
525
Th17 cells was reduced in the central nervous system (CNS) in C .rodentium immunized mice
526
[89].
527
In a recent study, Philips et al. have induced experimental autoimmune encephalomyelitis (EAE)
528
in mice with myelin antigen and demonstrated that DAB389IL-2 (denileukin diftitox;
529
ONTAK™)- a recombinant fusion toxin- targeting IL-2R bearing cells, suppressed the γσT cell
530
subpopulation in spinal cord lesions and subsequently overcome the disease [88] (Table 4).
(PPMS)
and
Progressive-Relapsing
MS
(PRMS).
Clinical
AC C
EP
TE D
M AN U
MS
SC
RI PT
509
24
ACCEPTED MANUSCRIPT
Recently, some drugs have been approved for the treatment of autoimmune diseases, but their
532
therapeutic efficacy is controversial. DNA vaccines are a novel method for treating autoimmune
533
diseases in a highly specific manner [88].
534
In some cases, administration of DNA vaccines against tumor cells may cause autoimmunity
535
which is a disadvantage to the application of DNA as a vaccine.
536
Actually there are several claims and counter claims in association with DNA vaccinations.
537
In 1997, Mor et al. analyzed the potential of DNA vaccines to induce or enhance autoimmunity
538
by administrating DNA vaccine to the mice. Normal mice immunized several times did not
539
develop clinical nor serological signs of autoimmune disease [90].
540
However, complementary investigations are needed for identifying the certain effect of DNA
541
vaccination on autoimmune disease.
542
The results obtained from experimental models of infectious diseases, cancer and
543
autoimmunity suggest that it is only the matter of time that clarifies the complete safety
544
and efficacy of DNA toxin vaccines. Until now, there are limited studies concerning DNA
545
vaccine associated with bacterial toxins and still a number of questions must be answered
546
to assess its complete safety and effectiveness for prevention and treatment of
547
aforementioned diseases.
548
7. Concluding remarks and future perspectives
549
The discovery of the conventional vaccines -first generation vaccines- was the most
550
important development in the last century which rescued millions of people from different
551
diseases. Subunit vaccines –second generation vaccines- developed to decrease the risks of
552
live attenuated vaccines and third generation vaccines namely DNA vaccines produced to
AC C
EP
TE D
M AN U
SC
RI PT
531
25
ACCEPTED MANUSCRIPT
overcome the shortage of other vaccines including problems in transportation and its high
554
production costs [91].
555
The most important feature of DNA vaccines is the ability to develop either humoral or
556
cellular immune response or both that makes them convenient for the treatment of
557
different diseases. In many cases of infectious diseases, DNA vaccine gives proper
558
protection against foreign antigens but only a limited number of vaccines exist for some
559
bacteria and the use of bacterial toxin sequences is somehow proved to be a useful
560
approach [91].
561
In cancer therapy, bacterial DNA toxin vaccines proved to have less side-effects and more
562
effective compared to conventional cancer therapy [92].
563
Findings about the treatment of autoimmune diseases using DNA vaccines are
564
controversial and bacterial toxins are less studied in this case and complementary studies
565
are needed.
566
All in all, DNA vaccine is a revolutionary technology in the field of preventive immunology,
567
still new approaches such as implementation of bacterial toxin sequences as an adjuvant
568
for optimization and augmentation of its immunogenicity is needed to be used.
569
On the other hand, we know that bacteria are not just single-celled organisms that cause diseases
570
in human and other animals and bacterial toxins are not toxic for all cells and the host organism.
571
In some cases toxin may adjust the function of the target cell and still little is known about their
572
mechanisms. Some toxins target the molecular machinery of the cell cycle. Therefore these toxin
573
producing genes can be implemented in DNA toxin vaccine development. But still, further
574
research for more complete understanding of the molecular action of bacterial toxins in vivo is
575
needed. It seems that some bacterial toxins play no role in pathogenesis and some have different
AC C
EP
TE D
M AN U
SC
RI PT
553
26
ACCEPTED MANUSCRIPT
effects on various cell types because although toxin molecules act in the same mode in different
577
cell types, the activated signaling pathways differ from one cell to another, which result in
578
various cellular consequences.
579
The characteristics of some toxins such as Pasteurella multocida toxin (PMT) and Cytotoxic
580
necrotizing factor 1 (CNF-1) produced by E. coli make them function as tumor inducers which
581
their properties must be studied carefully in vaccine development [93]. As some toxins are
582
effective for treating cancerous cells but exposing cells to the high quantity of toxins may have a
583
reverse effect on them. Low quantities might still induce signaling pathway, for this reason the
584
mode and amount of injection are of importance [94].
585
The DNA toxin vaccines needed more attention and the research on this area may bring
586
considerable promotions in curing cancer, autoimmune and infectious patients in the future. In
587
addition, some experimental and clinical evidences show the efficacy of engineered toxin
588
vaccines against certain tumors and infectious diseases.
589
In conclusion, DNA vaccines possess significant advantages including rapid manipulation, easy
590
translocation via plasmids, applicable as multifunctional vaccines, stability with the least
591
adverse effects in humans, cost-effectiveness and reliability which show a bright promising
592
future relating to DNA vaccines [95].
593
8. Funding
594
This study was not funded by any grant from institute.
595
9. Conflict of Interest
596
No conflict of interest declared.
597
10. Informed consent
598
All of the authors had the same contribution in the article and are agreed to submit manuscript.
AC C
EP
TE D
M AN U
SC
RI PT
576
27
ACCEPTED MANUSCRIPT
11. Figures:
600
Fig 1. Schematic representation of PAMPs recognition by TLRs and NLRs, respectively.
601
Figure 2. Schematic representation of immunological mechanism of DNA vaccine in
602
comparison with traditional vaccines.
603
Figure 3. Schematic representation of immunological mechanism of DNA vaccine in
604
respiratory infections.
605
Figure 4. Schematic representation of immunological mechanism of DNA vaccine in cancer
606
treatment.
SC
RI PT
599
M AN U
607 References
609
1.
Liu, M.A., DNA vaccines: a review. Journal of Internal Medicine, 2003. 253(4): p. 402-410.
610 611
2.
Liu, M.A., DNA vaccines: an historical perspective and view to the future. Immunological Reviews, 2011. 239(1): p. 62-84.
612 613
3.
Li, L., F. Saade, and N. Petrovsky, The future of human DNA vaccines. Journal of biotechnology, 2012. 162(2): p. 171-182.
614 615
4.
Mazid, R., M. X. Tan, and M. K. Danquah, Molecular Delivery of Plasmids for Genetic Vaccination. Current Pharmaceutical Biotechnology, 2013. 14(6): p. 615-622.
616 617
5.
618 619
6.
620
7.
AC C
EP
TE D
608
Fioretti, D., S. Iurescia, and M. Rinaldi, Recent advances in design of immunogenic and effective naked DNA vaccines against cancer. Recent Pat Anticancer Drug Discov, 2014. 9(1): p. 66-82.
Mellman, I. and R.M. Steinman, Dendritic Cells-Specialized and Regulated Antigen Processing Machines. Cell, 2001. 106(3): p. 255-258. Khan, K.H., DNA vaccines: roles against diseases. Germs, 2013. 3(1): p. 26.
28
ACCEPTED MANUSCRIPT
8.
Fynan, E.F., et al., DNA vaccines: A novel approach to immunization. International Journal of Immunopharmacology, 1995. 17(2): p. 79-83.
623 624
9.
Lowrie, D.B., et al., Towards a DNA vaccine against tuberculosis. Vaccine, 1994. 12(16): p. 15371540.
625 626
10.
Wolff, J.A. and V. Budker, The mechanism of naked DNA uptake and expression. Adv Genet, 2005. 54: p. 3-20.
627 628
11.
Yin, H., et al., Non-viral vectors for gene-based therapy. Nature Reviews Genetics, 2014. 15(8): p. 541-555.
629 630
12.
Hung, C.-F. and T.-C. Wu, Superior molecular vaccine linking the translocation domain of a bacterial toxin to an antigen. 2012, Google Patents.
631 632
13.
Sumithra, T.G., et al., Progress in DNA Vaccinology against Bacterial Diseases–An Update. DNA, 2013. 2013: p. 09-20.
633 634
14.
Yin, J.-x., et al., Pertussis toxin modulates microglia and T cell profile to protect experimental autoimmune encephalomyelitis. Neuropharmacology, 2014. 81: p. 1-5.
635 636
15.
Bolhassani, A. and S.R. Yazdi, DNA immunization as an efficient strategy for vaccination. Avicenna journal of medical biotechnology, 2009. 1(2): p. 71.
637 638
16.
Qiao, B., et al., Induction of systemic lupus erythematosus-like syndrome in syngeneic mice by immunization with activated lymphocyte-derived DNA. Rheumatology, 2005. 44(9): p. 1108-1114.
639 640
17.
Wassenaar, T.M. and W. Gaastra, Bacterial virulence: can we draw the line? FEMS microbiology letters, 2001. 201(1): p. 1-7.
641 642
18.
643 644
19.
Sansonetti, P.J. and J.P. Di Santo, Debugging how bacteria manipulate the immune response. Immunity, 2007. 26(2): p. 149-161.
645 646
20.
Freytag, L. and J. Clements, Bacterial toxins as mucosal adjuvants, in Defense of Mucosal Surfaces: Pathogenesis, Immunity and Vaccines. 1999, Springer. p. 215-236.
AC C
EP
TE D
M AN U
SC
RI PT
621 622
Oswald, E., et al., Bacterial toxins that modulate host cell-cycle progression. Current opinion in microbiology, 2005. 8(1): p. 83-91.
29
ACCEPTED MANUSCRIPT
21.
Eriksson, K. and J. Holmgren, Recent advances in mucosal vaccines and adjuvants. Current opinion in immunology, 2002. 14(5): p. 666-672.
649 650
22.
Arakawa, T., et al., A plant-based cholera toxin B subunit–insulin fusion protein protects against the development of autoimmune diabetes. Nature biotechnology, 1998. 16(10): p. 934-938.
651 652
23.
Hung, C.-F., et al., Cancer Immunotherapy Using a DNA Vaccine Encoding the Translocation Domain of a Bacterial Toxin Linked to a Tumor Antigen. Cancer Research, 2001. 61(9): p. 3698-3703.
653 654
24.
Curtiss, R., III, Bacterial infectious disease control by vaccine development. The Journal of Clinical Investigation, 2002. 110(8): p. 1061-1066.
655
25.
Tafalla, C., et al., 7 Adjuvants in Fish Vaccines. Fish Vaccination, 2014: p. 68.
656 657
26.
Pizza, M., et al., Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants. Vaccine, 2001. 19(17-19): p. 2534-41.
658 659 660
27.
Arrington, J., et al., Plasmid Vectors Encoding Cholera Toxin or the Heat-Labile Enterotoxin from Escherichia coli Are Strong Adjuvants for DNA Vaccines. Journal of Virology, 2002. 76(9): p. 4536-4546.
661 662
28.
Akira, S., S. Uematsu, and O. Takeuchi, Pathogen Recognition and Innate Immunity. Cell, 2006. 124(4): p. 783-801.
663 664 665
29.
Van De Kant, K., et al., Interaction Between Genetic Variants Of Cd14 And Toll-Like Receptors With Bacteria And Regulatory T-Cells In The Development Of Childhood Asthma. Am J Respir Crit Care Med, 2013. 187: p. A3521.
666 667
30.
Hossain, M.M. and M.-N. Norazmi, Pattern Recognition Receptors and Cytokines in Mycobacterium tuberculosis Infection—The Double-Edged Sword? BioMed research international, 2013. 2013.
668 669
31.
670 671
32.
AC C
EP
TE D
M AN U
SC
RI PT
647 648
Kanneganti, T.-D., M. Lamkanfi, and G. Núñez, Intracellular NOD-like receptors in host defense and disease. Immunity, 2007. 27(4): p. 549-559.
Kant, R., et al., Immunostimulatory CpG motifs in the genomes of gut bacteria and their role in human health and disease. Journal of medical microbiology, 2014. 63(Pt 2): p. 293-308.
30
ACCEPTED MANUSCRIPT
33.
Del Vecchio, M., et al., Interleukin-12: biological properties and clinical application. Clinical Cancer Research, 2007. 13(16): p. 4677-4685.
674 675
34.
Akira, S., Innate immunity and adjuvants. Philosophical Transactions of the Royal Society B: Biological Sciences, 2011. 366(1579): p. 2748-2755.
676 677
35.
Proft, T., Bacterial Toxins: Genetics, Cellular Biology and Practical Applications. 2013: Horizon Scientific Press.
678 679
36.
Mihret, A., The role of dendritic cells in Mycobacterium tuberculosis infection. Virulence, 2012. 3(7): p. 654-659.
680 681
37.
Zom, G.G., et al., 7 TLR Ligand—Peptide Conjugate Vaccines: Toward Clinical Application. Advances in immunology, 2012. 114: p. 177.
682 683
38.
McGuirk, P., S.C. Higgins, and K.H. Mills, The role of regulatory T cells in respiratory infections and allergy and asthma. Current allergy and asthma reports, 2010. 10(1): p. 21-28.
684 685 686
39.
Valitutti, S., et al., Sustained signaling leading to T cell activation results from prolonged T cell receptor occupancy. Role of T cell actin cytoskeleton. The Journal of experimental medicine, 1995. 181(2): p. 577-584.
687
40.
Hackett, C.J. and D.A. Harn, Vaccine Adjuvants. 2006: Springer.
688 689
41.
Mak, T.W. and M.E. Saunders, The immune response: basic and clinical principles. 2005: Academic Press.
690 691
42.
Pasetti, M.F., et al., Immunology of gut mucosal vaccines. Immunological reviews, 2011. 239(1): p. 125-148.
692 693
43.
694 695
44.
Nascimento, I.P. and L.C. Leite, Recombinant vaccines and the development of new vaccine strategies. Braz J Med Biol Res, 2012. 45(12): p. 1102-11.
696 697
45.
Ingolotti, M., et al., DNA vaccines for targeting bacterial infections. Expert Rev Vaccines, 2010. 9(7): p. 747-63.
AC C
EP
TE D
M AN U
SC
RI PT
672 673
Holmgren, J., et al., Mucosal immunisation and adjuvants: a brief overview of recent advances and challenges. Vaccine, 2003. 21 Suppl 2: p. S89-95.
31
ACCEPTED MANUSCRIPT
46.
Liu, M.A., DNA vaccines: an historical perspective and view to the future. Immunol Rev, 2011. 239(1): p. 62-84.
700 701 702
47.
Frahm, N., et al., CD8(+) T-cell Mediated HIV Inhibition after Vaccination with a DNA/Recombinant Ad5 (rAd5) HIV Vaccine Is Similar to that Seen in Treated HIV Infection. AIDS Res Hum Retroviruses, 2014. 30 Suppl 1: p. A84.
703 704
48.
Saltzman, W.M., H. Shen, and J.L. Brandsma, DNA vaccines: methods and protocols. Vol. 127. 2006: Springer.
705 706
49.
Akira, S. and H. Hemmi, Recognition of pathogen-associated molecular patterns by TLR family. Immunology letters, 2003. 85(2): p. 85-95.
707
50.
Kresina, T.F., An introduction to molecular medicine and gene therapy. 2001: Wiley Online Library.
708 709
51.
Patel, K.G., et al., Escherichia coli-based production of a tumor idiotype antibody fragment--tetanus toxin fragment C fusion protein vaccine for B cell lymphoma. Protein Expr Purif, 2011. 75(1): p. 15-20.
710 711 712
52.
Chudley, L., et al., DNA fusion-gene vaccination in patients with prostate cancer
713
53.
Steinhagen, F., et al., TLR-based immune adjuvants. Vaccine, 2011. 29(17): p. 3341-3355.
714 715
54.
Lahiri, A., P. Das, and D. Chakravortty, Engagement of TLR signaling as adjuvant: towards smarter vaccine and beyond. Vaccine, 2008. 26(52): p. 6777-6783.
716 717
55.
Fioretti, D., et al., DNA vaccines: developing new strategies against cancer. BioMed Research International, 2010. 2010.
718 719
56.
720 721
57.
M AN U
SC
RI PT
698 699
induces high-frequency CD8+ T-cell responses and increases PSA doubling time.
AC C
EP
TE D
Cancer Immunology, Immunotherapy, 2012. 61(11): p. 2161-2170.
Riedel, S., Edward Jenner and the history of smallpox and vaccination. Proc (Bayl Univ Med Cent), 2005. 18(1): p. 21-5. Berche, P., Louis Pasteur, from crystals of life to vaccination. Clinical Microbiology and Infection, 2012. 18(s5): p. 1-6.
32
ACCEPTED MANUSCRIPT
58.
Verardi, P.H., A. Titong, and C.J. Hagen, A vaccinia virus renaissance: new vaccine and immunotherapeutic uses after smallpox eradication. Human vaccines & immunotherapeutics, 2012. 8(7): p. 961.
725 726 727
59.
Myron M., L., Gordon Dougan, Michael F. Good, Margaret A. Liu, Gary J. Nabel, James P. Nataro, Rino Rappouli, New Generation Vaccines. 2010, New York: informa healthcare.
728 729
60.
Gupta, R.K., Aluminum compounds as vaccine adjuvants. Advanced drug delivery reviews, 1998. 32(3): p. 155-172.
730 731
61.
Dong, K., The Evolution and Value of Diphtheria Vaccine. KSBB Journal, 2011. 26(6): p. 491504.
732 733 734
62.
Rydell, N. and I. Sjöholm, Mucosal vaccination against diphtheria using starch microparticles as adjuvant for cross-reacting material (CRM197) of diphtheria toxin. Vaccine, 2005. 23(21): p. 27752783.
735 736
63.
Mäkelä, P.H., Vaccines, coming of age after 200 years. FEMS microbiology reviews, 2000. 24(1): p. 9-20.
737 738 739 740
64.
Vadesilho, C.F., et al., Characterization of the antibody response elicited by immunization with pneumococcal surface protein A (PspA) as recombinant protein or DNA vaccine and analysis of protection against an intranasal lethal challenge with< i> Streptococcus pneumoniae. Microbial pathogenesis, 2012. 53(5): p. 243-249.
741
65.
Sandhu, S.S., Recombinant DNA Technology. 2010: IK International Pvt Ltd.
742
66.
Pereira, V.B., et al., Use of bacteria in DNA vaccine delivery. vaccine, 2013. 12: p. 15.
743 744
67.
745 746 747
68.
AC C
EP
TE D
M AN U
SC
RI PT
722 723 724
DeLeo, F.R. and H.F. Chambers, Reemergence of antibiotic-resistant Staphylococcus aureus in the genomics era. The Journal of clinical investigation, 2009. 119(9): p. 2464-2474. Li, X., W. Xu, and S. Xiong, A novel tuberculosis DNA vaccine in an HIV-1 p24 protein backbone confers protection against Mycobacterium tuberculosis and simultaneously elicits robust humoral and cellular responses to HIV-1. Clinical and Vaccine Immunology, 2012. 19(5): p. 723-730.
33
ACCEPTED MANUSCRIPT
69.
Baliban, S.M., et al., An Optimized, Synthetic DNA Vaccine Encoding the Toxin A and Toxin B Receptor Binding Domains of Clostridium difficile Induces Protective Antibody Responses In Vivo. Infection and immunity, 2014. 82(10): p. 4080-4091.
751
70.
Alouf, J.E., The comprehensive sourcebook of bacterial protein toxins. 2005: Academic Press.
752 753
71.
Reichert, J.M. and J.B. Wenger, Development trends for new cancer therapeutics and vaccines. Drug discovery today, 2008. 13(1): p. 30-37.
754 755
72.
Mellman, I., G. Coukos, and G. Dranoff, Cancer immunotherapy comes of age. Nature, 2011. 480(7378): p. 480-489.
756
73.
Tew, K.D. and P . Fisher, Advances in Cancer Research. 2012: Academic Press.
757
74.
Emens, L.A., Cancer vaccines: on the threshold of success. 2008.
758
75.
Elledge, S., et al., Genetic Approaches to Cancer. The FASEB Journal, 2013. 27: p. 451.1.
759 760
76.
Rice, J., C.H. Ottensmeier, and F.K. Stevenson, DNA vaccines: precision tools for activating effective immunity against cancer. Nature Reviews Cancer, 2008. 8(2): p. 108-120.
761 762 763
77.
Rice, J., S. Buchan, and F.K. Stevenson, Critical components of a DNA fusion vaccine able to induce protective cytotoxic T cells against a single epitope of a tumor antigen. The Journal of Immunology, 2002. 169(7): p. 3908-3913.
764 765
78.
Stevenson, F.K., et al., DNA fusion vaccines enter the clinic. Cancer Immunology, Immunotherapy, 2011. 60(8): p. 1147-1151.
766 767
79.
768 769 770
80.
771 772
81.
AC C
EP
TE D
M AN U
SC
RI PT
748 749 750
Patyar, S., et al., Review Bacteria in cancer therapy: a novel experimental strategy. J. Biomed. Sci, 2010. 17(1): p. 21-30.
Chen, J., et al., Enhancement of Helicobacter pylori outer inflammatory protein DNA vaccine efficacy by co‐delivery of interleukin‐2 and B subunit heat‐labile toxin gene encoded plasmids. Microbiology and immunology, 2012. 56(2): p. 85-92. Weldon, J.E. and I. Pastan, A guide to taming a toxin–recombinant immunotoxins constructed from Pseudomonas exotoxin A for the treatment of cancer. FEBS Journal, 2011. 278(23): p. 4683-4700.
34
ACCEPTED MANUSCRIPT
82.
Yang, X., et al., Diphtheria Toxin–Epidermal Growth Factor Fusion Protein DAB389EGF for the Treatment of Bladder Cancer. Clinical Cancer Research, 2013. 19(1): p. 148-157.
775 776
83.
Gao, Z., et al., C terminus of Clostridium perfringens enterotoxin downregulates CLDN4 and sensitizes ovarian cancer cells to Taxol and Carboplatin. Clinical Cancer Research, 2011. 17(5): p. 1065-1074.
777
84.
Wayne, A.S., et al., Immunotoxins for leukemia. Blood, 2014. 123(16): p. 2470-2477.
778 779
85.
Denis-Mize, K.S., et al., Analysis of immunization with DNA encoding Pseudomonas aeruginosa exotoxin A. FEMS Immunol Med Microbiol, 2000. 27(2): p. 147-54.
780 781
86.
Calvo, A.C., et al., Fragment C of tetanus toxin: new insights into its neuronal signaling pathway. International journal of molecular sciences, 2012. 13(6): p. 6883-6901.
782
87.
Janeway, C.A., et al., Autoimmune responses are directed against self antigens. 2001.
783 784 785
88.
Phillips, S.M., et al., Suppression of murine experimental autoimmune encephalomyelitis by interleukin-2 receptor targeted fusion toxin, DAB< sub> 389 IL-2. Cellular immunology, 2010. 261(2): p. 144-152.
786 787 788
89.
Smith, A.J., et al., Comparison of a classical Th1 bacteria versus a Th17 bacteria as adjuvant in the induction of experimental autoimmune encephalomyelitis. Journal of neuroimmunology, 2011. 237(1): p. 33-38.
789 790
90.
Mor, G., et al., Do DNA vaccines induce autoimmune disease? Human gene therapy, 1997. 8(3): p. 293-300.
791 792 793
91.
Grunwald, T. and S. Ulbert, Improvement of DNA vaccination by adjuvants and sophisticated delivery devices: vaccine-platforms for the battle against infectious diseases. Clinical and experimental vaccine research, 2015. 4(1): p. 1-10.
794 795
92.
796 797 798
93.
AC C
EP
TE D
M AN U
SC
RI PT
773 774
Tiptiri-Kourpeti, A., et al., DNA vaccines to attack cancer: Strategies for improving immunogenicity and efficacy. Pharmacology & therapeutics, 2016. Oubrahim, H., et al., Pasteurella multocida toxin (PMT) upregulates CTGF which leads to mTORC1 activation in Swiss 3T3 cells. Cellular signalling, 2013. 25(5): p. 1136-1148.
35
ACCEPTED MANUSCRIPT
799 800
94.
Gardiner, D.F., et al., A DNA vaccine targeting the receptor-binding domain of Clostridium difficile toxin A. Vaccine, 2009. 27(27): p. 3598-3604.
801 802
95.
Saade, F. and N. Petrovsky, Technologies for enhanced efficacy of DNA vaccines. Expert review of vaccines, 2012. 11(2): p. 189-209.
AC C
EP
TE D
M AN U
SC
RI PT
803 804 805
36
ACCEPTED MANUSCRIPT
Table 1. Advantages and disadvantages of DNA vaccine vs. traditional vaccine
Safety
No risk of infection
Immunogenicity
Long lasting immunity Well tolerated No adverse effect
Cost
AC C
Spectrum
EP
TE D
Development Production
Thermal stability Longer life time Easy to store and transport Rapid to produce Can encodes for several antigens and Simple design with possibility of further modification Easy and large-scale production Relatively inexpensive to produce Several plasmids could be used to make a broad spectrum vaccine
M AN U
Stability
Disadvantages Induce immunologic tolerance Can affect the genes which control the growth of cells Induce antibody production Antibiotic resistance Relatively poor immunogenicity Not useful against non-protein based antigens Immunotoxicity
RI PT
Advantages
SC
Characteristics
-
-
Difficulty/cost of delivery Not effective for all common pathogens/diseases
ACCEPTED MANUSCRIPT
Adjuvant
Host
Refs.
Clostridium difficile
Alum compounds
Mice, Hamster
(70)
Clostridium tetani toxoid
Alum compounds
Human
(59)
Corynebacteriumdiphtheriae toxoid
Alum compounds
Human
(59)
Bordetellapertusis toxin
Alum compounds with others such as liposomes and monophosphoryl lipid A, Gamma Inulin Alhydrogel
Human
(59)
Mice
(71)
Mice
(13, 69)
Pseudomonas aeruginosa Exotoxin A
Vibrio cholerae toxin
T domain belonging to diphtheria toxin Cholera toxin b subunit (CTB) CT, IL-1α, LPS, CpG, Pam3CSK4, 3M-019, resiquimod/R848 or c48/80 HemolysinCyclodextrin complex CTB
TE D
Bacillus anthracis toxin
M AN U
Clostridium botulinum toxoid
Staphylococcus aureus α-toxin
AC C
EP
Escherrichia coli (ETEC) toxoid
RI PT
Infectious disease
SC
Table 2.DNA toxin vaccines for infectious diseases
Different animals (72) and Human Mice (73)
Mice
(69)
Human
(69)
ACCEPTED MANUSCRIPT
Table 3.DNA tetanus toxin vaccines for cancer therapy Cancer Genetic encoding
Host
Refs.
DNA Plasmid/HER2 ICD
Human
(54)
Cervical cancer
DNA Plasmid/mutated E7 HPV antigen
Human
(54)
Liver cancer
DNA Plasmid/Murine α-fetoprotein,
RI PT
Breast cancer
Human-
Adenoviral vector
Melanoma
DNA Plasmid/Synchrotope MA2M DNA Plasmid/pSEM Ex-DNA/rhPSA DNA Plasmid/PSMA DNA Plasmid/Bacterial HSP90
Prostate cancer Tumors
Human
SC
DNA idiotopicscFv and TTFrC
Mice
M AN U
Lymphatic cancer
Table 4.DNA toxin vaccines for autoimmune diseases therapy Host Allergy Vaccine elements
Adjuvant arthritis
(54)
Human
(54)
Mammals (91)
Refs.
Lewis Rat
(91)
Mycobacterial &Chlamydia HSP60 diphtheria toxin
Human
(91)
Mice
(93)
AC C
Multiple sclerosis (MS)
Human
Lewis Human
EP
Atherosclerosis
(54)
Mycobacteria and Escherichia coliHSP60& HSP70/plasmid MycobacteriaHSP60/plasmid
TE D
Rheumatoid arthritis
(54)
Rats, (91, 96)
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT