G Model
ARTICLE IN PRESS
JVAC 16556 1–9
Vaccine xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Vaccine journal homepage: www.elsevier.com/locate/vaccine
Review
1
Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines
2
3
4
Q1
a
5 6
Jorge A. Salazar-González a , Carlos Angulo b , Sergio Rosales-Mendoza a,∗ Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San
Q2 Luis Potosí 78210, SLP, Mexico b
Grupo de Inmunología y Vacunología, Centro de Investigaciones Biológicas del Noroeste, SC., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., C.P. 23096 Mexico City, Mexico
7 8 9
a r t i c l e
10 27
i n f o
a b s t r a c t
11
Article history: Received 3 May 2015 Received in revised form 25 May 2015 Accepted 28 May 2015 Available online xxx
12 13 14 15 16 17
26
Keywords: Oral vaccine Vaccine cost Transgenic plant Transient expression Transplastomic plant Edible crop E antigen Virus like particles
28
1. Introduction
18 19 20 21 22 23 24 25
29Q4 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45
Q3
Chikungunya virus is an emerging pathogen initially found in East Africa and currently spread into the Indian Ocean Islands, many regions of South East Asia, and in the Americas. No licensed vaccines against this eminent pathogen are available and thus intensive research in this field is a priority. This review presents the current scenario on the developments of Chikungunya virus vaccines and identifies the use of genetic engineered plants to develop attractive vaccines. The possible avenues to develop plantmade vaccines with distinct antigenic designs and expression modalities are identified and discussed considering current trends in the field. © 2015 Published by Elsevier Ltd.
Chikungunya virus (CHIKV) causes an infection typically characterized with fever, skin rash, incapacitating arthralgia, and severe synovitis [1]. This virus is transmitted by the Aedes mosquitoes and its name derives from the Swahili or Makonde word Kun qunwala that translates to “walk bent over”, which describes the posture of infected persons experiencing severe joint pain. Chikungunya is easily confused with dengue as they share the same vectors, symptoms, and geographical distribution; but differs in the absence of headache, and retro-orbital eye pain [2]. CHIKV, first isolated in 1952 from a febrile patient during an outbreak in the Makonde Plateau in the southern province of Tanzania (formerly Tanganyika), is a prevalent pathogen in tropical and subtropical regions of Africa, the Indian Ocean Islands, and south and southeast Asia among the Makonde tribe [2]. CHIKV is an enveloped alpha virus belonging to the family Togaviridae, whose genome consists of a single-stranded positivesense RNA of approximately 11.8 kb. The genome is capped at
∗ Corresponding author. Tel.: +52 444 826 2440; fax: +52 444 826 2440. E-mail address:
[email protected] (S. Rosales-Mendoza).
the 5 end and polyadenylated at the 3 end. The genomic structure of CHIKV encodes for the following: one open reading frame (5 ORF) yielding four non-structural proteins (nsP1–4) at the posttranslational level which participate in genome replication, RNA capping, polyprotein cleavage, and other functions required for viral replication; and another ORF that yields three major structural proteins (Capsid, E1, and E2) and two small cleavage products (E3 and 6K) [3]. The mature virion is 70 nm in diameter and contains 240 heterodimers of E2/E1 arranged as trimeric spikes on its surface. These heterodimer spikes are inserted into the plasma membrane of infected cells after transported through the secretory pathway. Cytoplasmic nucleocapsids containing the genomic RNA and 240 copies of the capsid protein bud from the cell surface to acquire the virion envelope and envelope protein spikes. The E1 and E2 glycoproteins form heterodimers that associate as trimeric spikes on the virion surface while E3 and 6K were demonstrated to act as helper proteins in the budding and maturation process of the virion envelope [4,5]. CHIKV is believed to be originated in Africa where two genetically distinct lineages have been identified: one containing all isolates from western Africa and the second comprising all southern and East African strains, as well as isolates from Asia. West African lineages caused multiple CHIKV epidemics in East Africa,
http://dx.doi.org/10.1016/j.vaccine.2015.05.104 0264-410X/© 2015 Published by Elsevier Ltd.
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68
G Model JVAC 16556 1–9 2 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101
102 103
104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
the Indian Ocean Islands, and many parts of South East Asia [6–11]. Before 2000 C.E., large outbreaks of CHIKV were rare but become more frequent afterwards. More recently new episodes of CHIKV have been reported in the Americas, further broadening the geographical spread of the disease, which has been associated with an emerging genetic variability [12,13] leading to hypotheses on possible mechanisms of evolutionary adaptation of the virus to the mosquito vector [14,15]. The main preventive strategy against Chikungunya is mosquito control, but this has proven to be difficult especially in poor countries; thus new strategies to fight the disease are needed. Currently the only treatments for CHIKV-disease symptoms are non-steroidal anti-inflammatory drugs. The re-emergence of CHIKV has led to the assessment of several potential treatments including ribavirin [16,17], chloroquine [18,19], and CHIKV antibodies [20–23]. In addition the peptides Latarcin (LATA) and Thanatin (THAN) as well as the protein PAP1, all of which having antiviral activity [24,25], have shown a promising potential to protect against CHIKV [26]. Among the explored strategies, vaccination is considered the ideal intervention to prevent the CHIKV infection; however no licensed vaccines for human use are available yet. Despite the development of several animal models, few of them have met the requirement to be used in pre-clinical studies to assess potential therapeutics. Recent epidemiological data showed the increasing importance of antibody-mediated protection against CHIKV [21–23], highlighting the feasibility of using anti-CHIKV antibodies as a passive immunotherapy or as a prophylactic treatment. However, information about the exact target of the adaptive immune response either in human or in animal models remains limited. In addition the cost for immunotherapies produced under conventional platforms should be considered, which is prohibitive for massive use in developing countries.
2. Immununopathogenesis of CHIKV infection and animal models Deciphering CHIKV specific molecular features and how the virus interacts with its host are key aspects to prevent, treat, or cure the infection. However, the knowledge of human CHIKV infection immunology is limited to small animal models (mouse) [27] in which muscle and joint disease were recently achieved in C57BL/6 mice [28,29]. Although the mouse model is useful at preclinical level for vaccine development, CHIKV disease mice models (young or immunodeficient mice) do not fully recapitulate human disease patterns in terms of infectivity and immune responses. Therefore, Labadie et al. [30] proposed a model for CHIKV infection in adult immunocompetent cynomolgus macaques (Macaca fascicularis). CHIKV pathogenesis using this animal model seems to resemble the viral, clinical, and immunopathological features observed in the human disease; and, interestingly, macrophages were identified as the main cellular reservoirs during the late stages of CHIKV infection in vivo. Overall, the inflammatory response to CHIKV infection in humans clearly contributes to virus elimination since the viral load has been associated to the serum levels of proinflamatory mediators such as IFN-alpha, IFN-gamma, IL-1RA, IL-6, MCP-1/CCL-2, IL-12, IP-10/CXCL-10, IL-18, and IL-18BP [31,32]. However it is important to point out that proinflamatory mediators are orchestrated and depend largely on the stage of the viral pathogenesis, as demonstrated in cynomolgus macaques after CHIKV experimental infection [30]. Nevertheless, the beneficial or deleterious effects of inflammation on viral persistence remain unclear even though CHIKV infection-associated markers have been described [33,34]. In general, T and B cells have been associated to the clearance of CHIKV since reduced immune
responses in mice models and aged NHPs promoted long-term virus persistence [35,36].
3. Production of vaccine candidates against CHIKV Based on the hypothesis that an efficacious CHIKV vaccine should resemble the viral infection to provide accurately immunoprotection against CHIKV disease [37], several promising vaccines have been recently evaluated. For example the immunization with virus-like particles (VLPs) in a monkey model elicited neutralizing antibodies against envelope proteins from different CHIKV strains, mediating protection against viremia when challenged with a high dose of CHIKV; moreover, the transfer of these antibodies into immunodeficient mice conferred protection against a subsequent lethal CHIKV challenge [38]. In addition, purifying human antiCHIKV antibodies from patients in the convalescent phase exhibit a high in vitro CHIKV neutralizing activity and a powerful prophylactic and therapeutic efficacy against CHIKV infection in a mouse model, which correlates with the fact that infected individuals are in general protected against reinfections [20]. Therefore, the protection against CHIKV disease is considered to be primarily mediated by humoral responses. This knowledge have supported the development of immunization approaches resembling the natural infection process as close as possible through the use attenuated CHIKV strains or VLPs, which mimic the infection mechanism and induce antibody-mediated. Among the technologies that have been explored for the development of CHIKV vaccines stand-out: formalin-inactivated viral vaccines [39,40], live-attenuated viruses [41–43], alpha virus chimeras [44–46], consensus-based DNA vaccines [47–50], and recently virus-like particle (VLP) vaccines and recombinant subunit vaccines. A detailed scenario on protein subunit vaccines development is provided below. CHIKV structural proteins form enveloped VLPs (eVLPs) when expressed alone in eukaryotic expression systems [51,52]. The first CHIKV eVLP-based vaccine candidate was reported in 2010 by researchers of the NIH and has become the most promising VLP-based vaccine against CHIKV. DNA transfection of a plasmid comprising the full-length CHIKV structural coding region C-E3E2-6K-E1 into human HEK293 cells successfully resulted in CHIKV VLPs assembly. The viral glycoproteins in the VLPs are organized in 240 E1–E2 heterodimers, which form 80 spikes on the VLP surface, resembling replication-competent alpha viruses. These eVLPs were isolated from the supernatant of transfected mammalian cells, purified, and used to immunize mice and nonhuman primates. Vaccination of rhesus macaques with 3 doses consisting of 20 g of eVLPs at 0, 4, and 24 weeks induced an antibody response that was sufficient to confer protection upon a high-dose CHIKV challenge 15 weeks after the last boosting [53]. These results demonstrated that immunization with these VLPs elicited neutralizing antibodies directed against envelope proteins and protected NHPs against a subsequent lethal CHIKV challenge, indicating a humoral-mediated mechanism of protection. The next developmental step for this vaccine consisted on performing a Phase I dose-escalation clinical trial under a 3-dose vaccination scheme (weeks 0, 4, and 20) of up to 40 g of eVLPs per administration. This vaccine was safe, well tolerated and immunogenic [38]. Another VLPs-based promising vaccine development has consisted on a measles vaccine expressing CHIKV VLPs. A single immunization with this vaccine fully protected mice from a lethal CHIKV challenge [54]. This vaccine induced high titers of neutralizing CHIKV antibodies although specific cellular immune responses were also elicited. CHIKV eVLPs have also been expressed in insect cells. Research performed since 2011 demonstrated that the expression of the structural coding regions C-E3-E2-6K-E1 in Sf21 insect cells led to the assembly of VLPs [55–57]. Interestingly, these eVLPs displayed
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
132 133
134
135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194
G Model JVAC 16556 1–9
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260
superior immunogenicity in subcutaneously immunized mice in comparison with their subunit counterparts and effectively protected against a CHIKV lethal challenge in both AG129 and WT mice models. Further analysis revealed an association between this protective effect and the neutralizing antibody responses, which were induced even for a single immunization scheme (1 g/mouse) without the need of co-administered adjuvants [56,57]. This technology was adopted by the pharmaceutical company Merck that conducted studies consisting on the immunization of Guinea pigs with two doses (on days 0 and 14) that ranged 0.01–10 g along with the Adju-Phos aluminum adjuvant. The induction of specific IgG neutralizing antibodies in a dose-dependent manner was observed [58]. Similar findings have been found using live-attenuated CHIKV vaccines, which have also been evaluated in mice models. For example, a single administration of a live-attenuated CHIKV vaccine (CHIKV/IRES) effectively activated T cells with a peak on day 10 post-immunization and elicited memory CD4+ and CD8+ T cells that produced IFN-␥, TNF-␣, and IL-2. However, only passive immunization with anti-CHIKV/IRES immune serum provided protection [59]. Another example is the CHIKV vaccine strain 181/clone25 (181/25) developed by the United States Army Medical Research Institute of Infectious Diseases (USAMRIID). It was demonstrated that a single intradermal footpad immunization of AG129 (defective in IFN-␣/ and IFN-␥ receptor signaling) or A129 (defective in IFN-␣/ receptor signaling) mice with the attenuated CHIK 181/25 vaccine resulted in different mortality rates. AG129 mice resulted in rapid mortality within 3–4 days while A129 mice survived even after wild type CHIKV-La Reunion challenge, with the associated induction of significant levels of IFN-␥, IL-12, and specific antibodies. This vaccine was well-tolerated and highly immunogenic in phase I and II clinical trials [60]. Overall, these data highlight the importance of IFNs and neutralizing antibody responses on the protection against CHIKV infection. Viral-vectored vaccines have also been developed against CHIKV. A novel CHIKV vaccine candidate called MVA-CHIKV was developed based on the highly attenuated modified vaccinia virus Ankara (MVA), which is a poxvirus vector. This vaccine candidate relies on the expression of the CHIKV C, E3, E2, 6K, and E1 structural genes and has proven to induce robust innate immune responses in human macrophages and monocyte-derived dendritic cells in terms of the IFN-, proinflammatory cytokines, and chemokines production. After adjuvant-free intraperitoneal immunization of C57BL/6 mice with the chimeric virus at 1 × 107 PFU doses, strong CHIKV-specific CD8+ T cell responses were induced. CHIKV-specific CD8+ T cells were preferentially directed against the E1 and E2 proteins. Neutralizing humoral responses were also induced at high titers. Remarkably, mice treated with a single dose of the MVACHIKV vaccine were fully protected from a CHIKV challenge [61]. Protein subunit vaccines based on portions of some viral proteins have also been generated. A formulation based on two components (the 254 aa C-terminus region of the E1 protein and the full-length E2 protein) was produced using Escherichia coli BL21 (DE3) as expression host. Subcutaneous immunization of BALB/c on days 0, 21, and 35 with 40 g of either truncated E1 or E2 using different adjuvants (Freund’s complete adjuvant, Alum, or Montanide ISA720) was performed. This subunit vaccine candidate was able to induce broad and long lasting neutralizing antibodies and cell-mediated immune responses [62]. Interestingly, at least two CHIKV sub-domains (E2 domain A and B) are associated with human protective immunity based on antibody-dependent neutralization [63]. Another subunit vaccine candidate against CHIKV is based on the 364 aa ectodomain from E2 produced in the E. coli BL21 (DE3) strain. The intramuscular administration of 50 g of the recombinant protein, delivered as liposomes (CadB), in BALB/c mice induced neutralizing IgG
3
antibodies and high levels of IFN-␥. A challenge assay with CHIKV was also conducted, observing complete protection in the vaccinated group whereas control mice exhibited high viral load in muscles, brain, spleen, and blood [39]. Epitope-based vaccines constitute the ideal vaccination approach as they can be designed to achieve proper immune responses in terms of breadth and neutralizing potential. However, this field has been rather narrowly explored in part due to the limited CHIKV B cell epitope mapping. Therefore, the identification of a wider set of CHIKV neutralizing epitopes is still a need for this field. One of the few investigations on this subject revealed 4 relevant epitopes in the E2 protein which were identified by their reactivity against sera from immunized mice. These epitopes include the previously reported E2EP3 epitope (STKDNFNVYKATRPYLAH, aa 2800–2818 of the polyprotein) and three new epitopes: KWQYNSPLVPRNAELGDRKGKIHIPFPLANVTCR (3033–3066 aa of the polyprotein), located in the acid-sensitive region (ASR); KKEVVLTVPTEGLEVTWGNNEPYKYW (3113–3138 aa of the polyprotein); and AGMCMCARRRCITPYELTPGATVPFL (3185 to 3210 aa of the polyprotein). All these epitopes cluster at the C-terminus of the E2 glycoprotein [49]. In fact Kam et al. [21,22] observed, using plasma samples from humans obtained during the early convalescent phase, that the naturally-acquired IgG response is dominated by neutralizing IgG3 antibodies which are mostly directed toward the single linear epitope E2EP3, a peptide located at the N-terminus of the E2 glycoprotein and exposed on the viral envelope. More recently, it was confirmed that epitopes on the exposed top-most and outer surfaces of the E2/E1 trimer structure may be useful for CHIKV neutralization by specific antibodies, whereas epitopes facing the interior of the trimer are not [64]. Overall, these findings pave the way for the development of CHIKV-specific multi-epitopic- or VLP-based vaccination strategies that can be efficiently produced in plant-based platforms. As mentioned above, the implementation of advanced animal models to test vaccine candidates at the preclinical level is of relevance, especially those that closely resemble the infection that occurs in humans. Due to the close lineage relationship between humans and macaques, macaque models of CHIKV infection have been developed [30,35,65,66]. These models allow comparing the adaptive immunity between humans and macaques. Therefore, sera from non-human primates infected with CHIKV could reveal new neutralizing epitopes. A remarkable research on mapping CHIKV epitopes drop a series of epitopes all along in the structural proteins in macaques and in humans, indicating that the E2EP3 and the 3025 to 3066 epitopes are recognized by both human and macaques; which is consistent with previous reports (Table 1 [67]). This active research path leading to new epitopes is opening new prospects for the development of epitope-based vaccines against CHIKV.
4. The plant-based vaccines scenario Current plant biotechnology tools make possible the use of plants as both efficient bio-factories and delivery vehicles of subunit vaccines [68]. A wide range of proteins of pharmaceutical interest have been expressed in plants with several plant-made vaccine candidates being evaluated in clinical trials, including those for swine influenza, rabies, and hepatitis B [69]. This technology offers substantial advantages such as the absence of mammalian pathogens in the production process, low production cost, no requirement for fermentation systems, and efficient synthesis of complex proteins [70,71]. It is also possible to design oral vaccines based on the induction of specific immune responses in the gut associated lymphoid tissues (GALT) by orally administering plant biomass from edible plant species. This oral administration
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309
310
311 312 313 314 315 316 317 318 319 320 321 322 323
G Model
ARTICLE IN PRESS
JVAC 16556 1–9
J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
4
Table 1 Comparison of human and macaque CHIKV B-cell epitopes (taken from Kam et al. [67]). CHIKV protein nsP1 nsP2 nsP3
nsP4 Capsid E3 E2
Identified B-cell epitope (human)
Amino acid
Identified B-cell epitope (macaque)
Amino acid
AEEEREAELTREALPPLQ
497–514
GVNSVAIPLLSTGVYSGG FGASSETFPITFGDFNEGEIESLSSELLT FGDFLPGEVDDLTDSDWSTCSCSD TDDELLDRAGGYIFS
1433–1450 1801–1867
IQMRTQVELLDEHISIDC TVPVAPPRRRRGRNLTVT
1489–1506 1729–1746
KDIVTKITPEGAEEW
2721–2735
AAIIQRLKRGCRLYLMSETPKVPTYR PPKKKPAQKKKKPGRRERMCMKIEND RPIFDNKGRVVAIVLGGA
1937–1962 2561–2586 2689–2706
LLQASLTCSPHRQRR STKDNFNVYKATRPYLAHC TDGTLKIQVSLQIGIKTDDSHDWTKLRY MDNHMPADAERAGL LTTTDKVINNCKVDQCHAAVTNHKKW HAAVTNHKKWQYNSPLVPRNAEL GDRKGKIHIPFPLAN PTVTYGKNQVIMLLYPDHPTLLSYRN PTEGLEVTWGNNEPYKYWPQLSTNGT LLSMVGMAAGMCMCARRRCITPY ELTPGATVPFL
2785–2799 2800–2818 2841–2882
STKDNFNVYKATRPYLAHC ATTEEIEVHMPPDTPDRT
2800–2818 2961–2978
GNVKITVNGQTVRYKCNC HAAVTNHKKWQYNSPLVPRNAEL GDRKGKIHIPFPLANVTCR
2985–3002 3025–3066
3009–3034 3025–3058 3073–3098 3121–3146 3177–3210
6K E1 Regions of B cell epitopes found that are common to both human and macaque are bold.
324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339
approach offers additional attractive features including friendly delivery, safer administration than parenteral vaccines, and the avoidance of purification [72]. Thus these vaccines are highly attractive vaccines due to their low production cost which will allow attaining proper vaccination coverage in developing countries. The production of functional immunogens in the plant cell has been reported thus far following either oral or parenteral immunization schemes. The distinct expression modalities to produce antigenic proteins in the plant cell are described in Table 2. Until now, highly efficient transient expression systems have been adopted by the industry to produce parenteral vaccines by purifying the antigen from plant tissues [73]. Therefore the concept of plant-based vaccines is in the transition from a vision to a reality, with several candidates under evaluation in clinical trials [69]. Table 3 lists the antigenic configurations successfully implemented in plant-based systems. The immunogenic activity of
subunit vaccines is aided by designs based on the B subunits of bacterial toxins [74,75], Virus Like Particles (VLPs) [76], or immunoglobulin-based immune complexes [77]. These approaches have allowed attaining high immunogenic activity for several vaccines administered by the oral, intranasal, and parenteral routes. The main antigenic configurations explored in plant-based vaccines are described below. 4.1. LTB- and CTB-based chimeras
340 341 342 343 344 345 346
347
The B subunits of either the cholera toxin (from Vibrio cholerae) or the heat labile toxin (from enterotoxigenic E. coli) called CTB and LTB, respectively; have played a relevant role on vaccinology since these toxin-derived molecules lack of toxic activity but retain the GM1 binding activity, show a high immunogenicity, and exert immunomodulatory effects toward co-administered or fused
Table 2 Characteristics of the expression modalities for plant-based vaccines. Expression modality
Advantages
Limitations
Possible CHIKV expression targets
References
Nuclear stable transformation
Stable transgene insertion Post-translational modifications Well established transformation methods
C, E3, E2, 6K, E1 nsP1–4
[110]
Nuclear transient expression
Short production time Very high levels of recombinant protein
C, E3, E2, 6K, E1
[111]
Chloroplast transformation
High levels of recombinant protein Polycistronic expression is viable Improved biosafety as transgene is inherited maternally Site-specific insertion through recombination Non-susceptible to gene silencing
Low levels of recombinant protein Non-site specific transgene insertion Horizontal gene transfer is possible Susceptible to gene silencing Low reproducibility Transient expression limit the Purification of the antigen is required Lack of complex post-translational modifications Long time for generation of transformants Limited transformation protocols
C, E3, 6K nsP1–4
[112]
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
348 349 350 351 352 353
G Model
ARTICLE IN PRESS
JVAC 16556 1–9
J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx Table 3 Antigen configuration on plant-made vaccines. Antigen configuration
Advantages
Disadvantages
References
CTB/LTB chimeras
High mucosal immunogenicity High immunogenicity
May induce tolerance
[113]
Limitations on the size of heterologous sequences in chimeric VLPs Depend of a high-yield protein expression The longest ELP subunits the easiest to purify, but also achieves low expression levels
[114]
VLPs
Immune complexes
ELPylation
High immunogenicity
High immunogenicity Robust expression Easy purification
[93]
[97]
368
unrelated antigens [78]. In the field of plant-based vaccines, these molecules have been extensively used as adjuvant carriers, especially within the goal of developing oral vaccines since CTB/LTB serve as transmucosal carriers able to deliver the antigen at the sub mucosa [79]; where they can be efficiently processed by dendritic cells (DC) with the subsequent induction of robust mucosal and systemic Th1 immune responses. Several vaccination models based on this approach have proven to be effective at the preclinical level [80] and a clinical trial was performed with potatoes expressing LTB with positive outcomes in terms of seroconversion [81]. Therefore, fusing specific epitopes or domains from protective CHIKV antigens to CTB/LTB will serve as an attractive approach in the development of mucosal vaccines produced in plant tissues. The use of edible plant species will greatly facilitate the development of low cost oral vaccine prototypes.
369
4.2. Virus-like particles
354 355 356 357 358 359 360 361 362 363 364 365 366 367
370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395
Virus-Like Particles (VLPs) are self-assembled structures derived from viral antigens that mimic the native architecture of viruses but lack the viral genome and thus are not infective. Several reports on the expression of properly assembled VLPs at adequate levels have been published for Influenza virus, Human papillomavirus, Human immunodeficiency virus, Norwalk virus, and Hepatitis B virus; among others (reviewed in [71]). Since recent reports on CHIKV eVLPs production in mammalian and bacterial expression systems have proven to be efficacious (see above), the adoption of this concept in the form of plant-derived vaccines is a possibility. The glycosylation patterns of VLP proteins have a major impact on their structure and function since these viral glycoproteins localize, guide, and potentiate the process of enveloped virus assembly. Therefore, the design of plant-made vaccines based on the E1 and E2 glycoproteins demands a platform with the capacity to perform these complex post-translational modifications. The N-glycan synthesis in the endoplasmic reticulum is relatively well conserved in eukaryotes [82], thus it is expected that plants will provide the functional machinery to produce CHIKV eVLPs properly. In fact, some differences on glycosylation are expected in plants in comparison to mammal cells; however, this may account for the antigenicity of the plant-made CHIKV antigen due to a better recognition by antigen presenting cells through pattern-recognition receptors. If the hypothesis of higher antigenicity due to plant glycosylation fails, an alternative path consists on using the glycoengineering strategies that have been implemented in plants through knocking out
5
or silencing by RNAi, the genes or transcripts coding for the target glycosyltransferases, and/or introducing mammalian glycosylation genes [83,84]. Therefore, the precedents on VLPs production in plant systems for targeting other pathogens make these systems a reliable path to develop attractive CHIKV vaccine candidates. 4.3. Immune complexes Plants can express complex recombinant proteins requiring post-translational modifications, which are problematic to generate using other production platforms such as bacterial systems [85]. It has long been known that primary and secondary antibody responses to model antigens can be enhanced by immunization with immune complexes (ICs) [86,87]. Importantly, it is well established that ICs can be cross-presented by antigen-presenting cells (APC) and can stimulate potent T cell responses via MHC class I as well as class II molecules [88,89]. It has also been suggested that, through binding to Fc receptors and complement receptors, the ICs localize on the surface of follicular dendritic cells (FDCs), which play an important role in the selection and affinity maturation of B cells, or the complexes might directly stimulate B cells via their complement receptors [90]. However the conventional preparation of ICs is not feasible for vaccine production at a commercial scale, since it relies on the use of either polyclonal antisera or expensive monoclonal antibodies cocktails (mAbs) to achieve multimerization with a given antigen. To address these limitations, Chargelegue et al. described, for the first time, the production of recombinant ICs in transgenic tobacco plants by expressing the tetanus toxin fragment C fused to a mAb. The design of the recombinant IC fusion molecule resulted in the expression of the antibody-antigen at a 1:2 ratio (an antigen molecule was fused to each antibody heavy chain). Moreover, it was demonstrated that serum antibody responses leading to protective immunity were induced without the use of additional adjuvants in subcutaneously immunized mice [91]. Using similar approaches, multiple antigens have been expressed in tobacco being able to induce both humoral and cellular immune responses that protected against intracellular pathogens such as Mycobacterium tuberculosis. Intranasal immunization of mice with ICs boosted the Bacillus Calmette–Guerin strain vaccine (BCG)induced immunity and, importantly, conferred further protection against M. tuberculosis infection [92]. Remarkably, these methodologies have been applied to target viruses. For example, transient expression of Ebola-based immune complexes in plants has been achieved by fusing the Ebola GP1 glycoprotein subunit to a specific humanized heavy chain of 6D8 IgG monoclonal antibody. Mice subcutaneously immunized with Ebola ICs developed high titers of Ebola-specific IgG antibodies [93]. Thus, these studies show that ICs might represent an attractive immunization strategy against viral antigens and represent a promise for the development of highly immunogenic CHIKV vaccines. 4.4. Elastin-like polypeptide fusions The major cost in the production of subunit vaccines is related to the separation and purification steps, which can account for up to 90% of total production costs [94]. A benefit of plant-based expression systems is that they allow for alternative purification methods, such as elastin-like polypeptide (ELP) fusion technology. The ELPs ***Val-Pro-Gly-Gly, Val-Pro-Gly-Val-Gly, Ala-Pro-Gly-Val-Gly-Val, and Val-Pro-Gly-Xaa-Gly)n (where Xaa represents one random amino acid excluding Pro) exhibit the unique characteristic of reversible phase transition, meaning that they can be precipitated out of solution and re-suspended again through temperature manipulation. The temperature-dependent, reversible aggregation/precipitation properties of ELPs provides an alternative to the
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
396 397 398 399 400
401
402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444
445
446 447 448 449 450 451 452 453 454 455 456 457
G Model JVAC 16556 1–9 6
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
Fig. 1. Schematic representation of CHIKV immunopathology and the implications of plant-based vaccines in preventing the CHIKV infection. (A) It is known that CHIKV activates immune mechanisms that include the recruitment of macrophages as well as lymphocytes, early secretion of IgG3, and secretion of IFN␣/. (B) Plant-based vaccines are proposed as a convenient approach to produce antigenic proteins in the form of immune complexes, multiepitope vaccines, or Virus like particles. These systems will allow for the evaluation of prime-boost immunization schemes against CHIKV, where plant-made antigens can be purified and used for parenteral priming while minimally processed plant material can be used for oral boosting.
458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479
480 481
482 483 484 485 486 487
cost-intensive affinity chromatography and allows to separate the protein of interest from host proteins via centrifugation of crude extracts [95]. The efforts on plant-derived vaccines against human diseases using this approach are limited. One of the few studies comprises the M. tuberculosis antigens Ag85B and ESAT-6, which were fused to ELP and expressed in transgenic tobacco [96]. The subcutaneous immunization of mice with either 5 g or 10 g of ICs on days 0, 14, and 28; induced long-lasting humoral immune responses after 84 days. Interestingly the ELP was not removed from the antigen and it was found that the ELP had no effect on the immune response against the target antigens in mice. Another interesting approach consisted on the production of ELP fused to an established soluble trimer-forming H5N1 HA (ELPylated H5 HA, or H5-ELP) in tobacco. These recombinant proteins were easily purified by the inverse transition cycling technique. The subcutaneous administration in BL6 mice induced neutralizing antibodies after two doses administered on days 0 and 14. It was also found that ELPylation does not interfere with the immunogenicity of the vaccine candidate [97]. Therefore, this precedent of positive outcomes on targeting an enveloped virus through the ELP technology represents a positive prospect for proposing CHIKV vaccine candidates under this configuration.
5. Prospects for using plant-based CHIKV immunization approaches Plant-based technologies offer a myriad of possibilities to develop new vaccines against CHIKV thanks to the improvements of the technology achieved during the last two decades. Since several subunit vaccines against CHIKV have been explored using other expression systems, there is sufficient knowledge to support a straightforward design of plant-based vaccines (Fig. 1). The
recent identification and characterization of linear CHIKV B cellepitopes will allow designing innovative vaccines. Multiepitope vaccines against CHIKV are also a possibility when producing plantbased vaccines since multiepitope chimeric proteins have been produced in plants leading to promising findings [98,99]. Our group has developed plant-made vaccines against HIV, which were based on multiepitope polypeptides and against Taenia solium, which were based on several peptides produced through the 2A-based ribosomal skip mechanism [100]. Other groups have developed multicomponent vaccines against enteric pathogens based on chimeric proteins [101]. Since some targets have been identified as protective antigens against CHIKV, the plants producing those antigens along with convenient carriers could be evaluated as CHIKV vaccine candidates. The vaccine developmental steps would comprise: (i) designing immunogens based on proteins or epitopes associated with immunoprotection against CHIKV and developing transgenic plants carrying the corresponding genes; (ii) characterizing the plant-made antigen in terms of yields and antigenic activity; (iii) proving the immunogenic activity at the preclinical level through test animals immunization under distinct administration routes, and (iv) planning clinical trials once efficacy and safety in test animals have been proven. Therefore, this scenario indicates that a new avenue on developing anti-CHIKV vaccines will be viable to implement with the experience gained in the field working with other pathogens over the last two decades. This will constitute a relevant research field, allowing for the development of low cost and efficient vaccines. However it should be recognized that the progress in the field of plant-made vaccines has been focused to parenteral formulations using transient expression systems [71], while oral vaccination approaches still require optimization to address proper immunogenicity, a better control on dosage as well as antigen degradation,
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520
G Model JVAC 16556 1–9
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
533
and finding a proper regulatory framework [102,103]. Regarding oral vaccines, a recently proposed idea is based on the inducible expression of antigens mediated by viral vectors in edible plant species; which will allow acquiring high expression levels avoiding the purification steps. Therefore, plant biomass expressing high levels of the antigen might be used for the formulation of low cost oral vaccines consisting of freeze-dried plant material delivered in gelatin capsules [71]. Overall a convenient possibility for immunization schemes against CHIKV infection comprises parenteral priming with purified plant-made antigens and subsequent oral boosts with freeze-dried plant material expressing the candidate subunit vaccine (Fig. 1), as it has been proposed for the case of Hepatitis B Virus infection [104].
534
6. Concluding remarks
521 522 523 524 525 526 527 528 529 530 531 532
559
Considering that no vaccines against CHIKV approved for human use are available and that the disease is already present in the Americas, the need for advancing in this research field is great. The following years will be critical to envision the real potential to address this issue. Considering that promising candidates are in preclinical trials, the implementation in parallel of low cost production platforms will be determinant as CHKV is mainly affecting developing countries where access to vaccine is limited due to the high cost of conventional vaccines. In this review, plantbased platforms have been identified as promising approaches to fight CHIKV. The maturation that this technology achieved during the last decade currently allows for the production of several vaccine types ranging from oral formulations, which will required long-term research to be assessed and optimized, to parenteral vaccines produced under GMP-compliant transient expression systems [105,106]. The latter has the best potential to become a reality in the near future. Based on previous experiences, such as the developments on influenza and other viral pathogens vaccines [107], VLPs are the most efficacious approach to render immunogenic formulations even at low doses [108]. For example, influenza plantbased vaccines are in an advanced developmental stage, being evaluated in clinical trials [109]. In conclusion, the concept of plant-based vaccines constitutes a relevant path for the development of CHIV vaccines with attractive features.
560
Acknowledgements
535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558
Current investigations from the group are supported by 562 Q5 CONACYT/México (grant CB-2008-01, 102109to SRM and grant CB563 2010-01, 151818 to CA) and FAI/UASLP/2015 to SRM. 561
564
565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584
References [1] Brighton SW, Simson IW. A destructive arthropathy following Chikungunya virus arthritis—a possible association. Clin Rheumatol 1984;3(2):253–8. [2] Ross RW. The Newala epidemic. III. The virus: isolation, pathogenic properties and relationship to the epidemic. J Hyg (London) 1956;54(2):177–91. [3] Strauss JH, Strauss EG. The alphaviruses: gene expression, replication, and evolution. Microbiol Rev 1994;58:491–562. [4] Kuhn RJ. Togaviridae: the viruses and their replication. In: Knipe DM, Howley PM, editors. Fields’ virology. 5th ed. NY, USA: Lippincott, Williams and Wilkins; 2007. p. 1001–22. [5] Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, et al. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 2010;468:709–12. [6] Munasinghe DR, Amarasekera PJ, Fernando CF. An epidemic of dengue-like fever in Ceylon (chikungunya—a clinical and haematological study). Ceylon Med J 1966;11:129–42. [7] Pavri K. Disappearance of Chikungunya virus from India and South East Asia. Trans R Soc Trop Med Hyg 1986;80(491). [8] Lam SK, Chua KB, Hooi PS, Rahimah MA, Kumari S, et al. Chikungunya infection—an emerging disease in Malaysia. Southeast Asian J Trop Med Public Health 2001;32:447–51.
7
[9] Renault P, Solet JL, Sissoko D, Balleydier E, Larrieu S, et al. A major epidemic of Chikungunya virus infection on Reunion Island, France, 2005–2006. Am J Trop Med Hyg 2007;77:727–31. [10] Duong V, Andries AC, Ngan C, Sok T, Richner B, et al. Reemergence of Chikungunya virus in Cambodia. Emerg Infect Dis 2012;18:2066–9. [11] Ansumana R, Jacobsen KH, Leski TA, Covington AL, Bangura U, et al. Reemergence of Chikungunya virus in Bo, Sierra Leone. Emerg Infect Dis 2013;19:1108–10. [12] Leparc-Goffart I, Nougairede A, Cassadou S, Prat C, de Lamballerie X. Chikungunya in the Americas. Lancet 2014;383:514. [13] Fischer M, Staples E. Update on emerging infections: news from the Centers for Disease Control and Prevention. Notes from the field: Chikungunya virus spreads in the Americas-Caribbean and South America, 2013–2014. Ann Emerg Med 2014;64(5):552–3. [14] Tsetsarkin KA, Vanlandingham DL, McGee CE, Higgs S. A single mutation in Chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog 2007;3:1895–906. [15] Schuffenecker I, Iteman I, Michault A, et al. Genome microevolution of Chikungunya viruses causing the Indian Ocean outbreak. PLoS Med 2006;3:1058–70. [16] Ravichandran R, Manian M. Ribavirin therapy for Chikungunya arthritis. J Infect Dev Ctries 2008;2:140–2. [17] Briolant S, Garin D, Scaramozzino N, Jouan A, Crance JM. In vitro inhibition of Chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res 2004;61:111–7. [18] De Lamballerie X, Boisson V, Reynier JC, Enault S, Charrel RN, Flahault A, et al. On chikungunya acute infection and chloroquine treatment. Vector Borne Zoonotic Dis 2008;8:837–9. [19] Khan M, Santhosh SR, Tiwari M, Lakshmana Rao PV, Parida M. Assessment of in vitro prophylactic and therapeutic efficacy of chloroquine against Chikungunya virus in vero cells. J Med Virol 2010;82:817–24. [20] Couderc T, Khandoudi N, Grandadam M, Visse C, Gangneux N, Bagot S, et al. Prophylaxis and therapy for Chikungunya virus infection. J Infect Dis 2009;200:516–23. [21] Kam YW, Lum FM, Teo TH, Lee WW, Simarmata D, Harjanto S, et al. Early neutralizing IgG response to Chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol Med 2012;4:330–43. [22] Kam YW, Lee WW, Simarmata D, Harjanto S, Teng TS, Tolou H, et al. Longitudinal analysis of the human antibody response to Chikungunya virus infection: implications for serodiagnosis and vaccine development. J Virol 2012;86(Dec (23)):13005–15. [23] Lum FM, Teo TH, Lee WW, Kam YW, Rénia L, Ng LF. An essential role of antibodies in the control of Chikungunya virus infection. J Immunol 2013;190:6295–302. [24] Aron GM, Irvin JD. Inhibition of herpes simplex virus multiplication by the pokeweed antiviral protein. Antimicrob Agents Chemother 1980;17:1032–3. [25] Ishag HZ, Li C, Huang L, Sun MX, Ni B, Guo CX, et al. Inhibition of Japanese encephalitis virus infection in vitro and in vivo by pokeweed antiviral protein. Virus Res 2013;171:89–96. [26] Rothan HA, Bahrani H, Shankar EM, Rahman NA, Yusof R. Inhibitory effects of a peptide-fusion protein (Latarcin-PAP1-Thanatin) against Chikungunya virus. Antiviral Res 2014;108:173–80. [27] Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, et al. A mouse model for Chikungunya: young age and inefficient Type-I interferon signaling are risk factors for severe disease. PLoS Pathog 2008;4:e29. [28] Gardner J, Anraku I, Le TT, Larcher T, Major L, et al. Chikungunya virus arthritis in adult wild-type mice. J Virol 2010;84:8021–32. [29] Morrison TE, Oko L, Montgomery SA, Whitmore AC, Lotstein AR, et al. A mouse model of Chikungunya virus-induced musculoskeletal inflammatory disease: evidence of arthritis, tenosynovitis, myositis, and persistence. Am J Pathol 2011;178:32–40. [30] Labadie K, Larcher T, Joubert C, Mannioui A, Delache B, Brochard P, et al. Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Invest 2010;120:894–906. [31] Chow A, Her Z, Ong EK, Chen JM, Dimatatac F, et al. Persistent arthralgia induced by Chikungunya virus infection is associated with interleukin6 and granulocyte macrophage colony-stimulating factor. J Infect Dis 2011;203:149–57. [32] Chirathaworn C, Rianthavorn P, Wuttirattanakowit N, Poovorawan Y. Serum IL-18 and IL-18BP levels in patients with Chikungunya virus infection. Viral Immunol 2010;23:113–7. [33] Hoarau JJ, Jaffar Bandjee MC, Krejbich Trotot P, Das T, Li-Pat-Yuen G, Dassa B, et al. Persistent chronic inflammation and infection by Chikungunya arthritogenic alphavirus in spite of a robust host immune response. J Immunol 2010;184:5914–27. [34] Dupuis-Maguiraga L, Noret M, Brun S, Le Grand R, Gras G, Roques P. Chikungunya disease: infection-associated markers from the acute to the chronic phase of arbovirus-induced arthralgia. PLoS Negl Trop Dis 2012;6:e1446. [35] Messaoudi I, Vomaske J, Totonchy T, Kreklywich CN, Haberthur K, Springgay L, et al. Chikungunya virus infection results in higher and persistent viral replication in aged rhesus macaques due to defects in anti-viral immunity. PLoS Negl Trop Dis 2013;7:e2343. [36] Hawman DW, Stoermer KA, Montgomery SA, Pal P, Oko L, Diamond MS, et al. Chronic joint disease caused by persistent Chikungunya virus infection is controlled by the adaptive immune response. J Virol 2013;87:13878–88.
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670
G Model JVAC 16556 1–9 8 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
[37] Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2010;11:1087–101. [38] Akahata W, Yang ZY, Andersen H, Sun S, Holdaway HA, Kong WP, et al. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med 2010;16:334–8. [39] White A, Berman S, Lowenthal JP. Comparative immunogenicities of Chikungunya vaccines propagated in monkey kidney monolayers and chick embryo suspension cultures. Appl Microbiol 1972;23:951–2. [40] Kumar M, Sudeep AB, Arankalle VA. Evaluation of recombinant E2 proteinbased and whole-virus inactivated candidate vaccines against Chikungunya virus. Vaccine 2012;30:6142–9. [41] Levitt NH, Ramsburg HH, Hasty SE, Repik PM, Cole Jr FE, Lupton HW. Development of an attenuated strain of Chikungunya virus for use in vaccine production. Vaccine 1986;4:157–62. [42] McClain DJ, Pittman PR, Ramsburg HH, Nelson GO, Rossi CA, Mangiafico JA, et al. Immunologic interference from sequential administration of live attenuated alphavirus vaccines. J Infect Dis 1998;177:634–41. [43] Edelman R, Tacket CO, Wasserman SS, Bodison SA, Perry JG, Mangiafico JA. Phase II safety and immunogenicity study of live Chikungunya virus vaccine TSI-GSD-218. Am J Trop Med Hyg 2000;62:681–5. [44] Wang E, Volkova E, Adams AP, Forrester N, Xiao SY, Frolov I, et al. Chimeric alphavirus vaccine candidates for chikungunya. Vaccine 2008;26:5030–9. [45] Wang D, Suhrbier A, Penn-Nicholson A, Woraratanadharm J, Gardner J, Luo M, et al. A complex adenovirus vaccine against Chikungunya virus provides complete protection against viraemia and arthritis. Vaccine 2011;29:2803–9. [46] Darwin JR, Kenney JL, Weaver SC. Transmission potential of two chimeric Chikungunya vaccine candidates in the urban mosquito vectors, Aedes aegypti and Ae. albopictus. Am J Trop Med Hyg 2011;84(6):1012–5. [47] Muthumani K, Lankaraman KM, Laddy DJ, Sundaram SG, Chung CW, Sako E, et al. Immunogenicity of novel consensus-based DNA vaccines against Chikungunya virus. Vaccine 2008;26:5128–34. [48] Mallilankaraman K, Shedlock DJ, Bao H, Kawalekar OU, Fagone P, Ramanathan AA, et al. A DNA vaccine against Chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl Trop Dis 2011;5(1):e928. [49] Hallengärd D, Lum FM, Kümmerer BM, Lulla A, Lulla V, García-Arriaza J, et al. Prime-boost immunization strategies against Chikungunya virus. J Virol 2014;88:13333–43. [50] Hallengärd D, Kakoulidou M, Lulla A, Kümmerer BM, Johansson DX, Mutso M, et al. Novel attenuated Chikungunya vaccine candidates elicit protective immunity in C57BL/6 mice. J Virol 2014;88:2858–66. [51] Bredenbeek PJ, Rice CM. Animal RNA virus expression systems. Sem Virol 1992;3:297–310. [52] Frolov I, Hoffman TA, Prágai BM, Dryga SA, Huang HV, Schlesinger S, et al. Alphavirus-based expression vectors: strategies and applications. Proc Natl Acad Sci USA 1996;93:11371–7. [53] Chang LJ, Dowd KA, Mendoza FH, Saunders JG, Sitar S, Plummer SH, et al. Safety and tolerability of Chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet 2014;384:2046–52. [54] Brandler S, Ruffié C, Combredet C, Brault JB, Najburg V, Prevost MC, et al. A recombinant measles vaccine expressing Chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with Chikungunya virus. Vaccine 2013;31:3718–25. [55] Metz SW, Geertsema C, Martina BE, Andrade P, Heldens JG, van Oers MM, et al. Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells. Virol J 2011;8:353. [56] Metz SW, Martina BE, van den Doel P, Geertsema C, Osterhaus AD, Vlak JM, Pijlman GP. Chikungunya virus-like particles are more immunogenic in a lethal AG129 mouse model compared to glycoprotein E1 or E2 subunits. Vaccine 2013;31:6092–6. [57] Metz SW, Gardner J, Geertsema C, Le TT, Goh L, Vlak JM, et al. Effective Chikungunya virus-like particle vaccine produced in insect cells. PLoS Negl Trop Dis 2013;7:e2124. [58] Wagner JM, Pajerowski JD, Daniels CL, McHugh PM, Flynn JA, Balliet JW, et al. Enhanced production of Chikungunya virus-like particles using a high-pH adapted spodoptera frugiperda insect cell line. PLoS ONE 2014;9:e94401. [59] Chu H, Das SC, Fuchs JF, Suresh M, Weaver SC, Stinchcomb DT, et al. Deciphering the protective role of adaptive immunity to CHIKV/IRES a novel candidate vaccine against Chikungunya in the A129 mouse model. Vaccine 2013;31:3353–60. [60] Partidos CD, Weger J, Brewoo J, Seymour R, Borland EM, Ledermann JP, et al. Probing the attenuation and protective efficacy of a candidate Chikungunya virus vaccine in mice with compromised interferon (IFN) signaling. Vaccine 2011;29:3067–73. [61] García-Arriaza J, Cepeda V, Hallengärd D, Sorzano CÓ, Kümmerer BM, Liljeström P, et al. A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against Chikungunya infection. J Virol 2014;88:3527–47. [62] Khan M, Dhanwani R, Rao PV, Parida M. Subunit vaccine formulations based on recombinant envelope proteins of Chikungunya virus elicit balanced Th1/Th2 response and virus-neutralizing antibodies in mice. Virus Res 2012;167:236–46. [63] Lee CH, Kam YW, Fric J, Malleret B, Koh E, Prakash C, et al. Chikungunya virus neutralization antigens and direct cell-to-cell transmission are revealed by human antibody-escape mutants. PLoS Pathog 2011;7:e1002390.
[64] Fong RH, Banik SS, Mattia K, Barnes T, Tucker D, Liss N, et al. Exposure of epitope residues on the outer face of the Chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014;88:14364–79. [65] Chen CI, Clark DC, Pesavento P, Lerche NW, Luciw PA, Reisen WK, et al. Comparative pathogenesis of epidemic and enzootic Chikungunya virus in a pregnant Rhesus macaque model. Am J Trop Med Hyg 2010;83: 1249–58. [66] Higgs S, Ziegler SA. A nonhuman primate model of Chikungunya disease. J Clin Invest 2010;120:657–60. [67] Kam YW, Lee WW, Simarmata D, Le Grand R, Tolou H, Merits A, et al. Unique epitopes recognized by antibodies induced in Chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS ONE 2014;9:e95647. [68] Govea-Alonso DO, Rybicki E, Rosales-Mendoza S. Plant-based vaccines as a global vaccination approach: current perspectives. In: Genetically engineered plants as a source of vaccines against wide spread diseases diseases—an integrated view. New York, NY: Springer; 2014. p. 265–80. [69] Yusibov V, Streatfield SJ, Kushnir N. Clinical development of plant-produced recombinant pharmaceuticals: vaccines, antibodies and beyond. Hum Vaccin 2011;7:313–21. [70] Govea-Alonso DO, Cardineau GA, Rosales-Mendoza S. Principles of plantbased vaccines. In: Genetically engineered plants as a source of vaccines against wide spread diseases—an integrated view. New York, NY: Springer; 2014. p. 1–14. ˜ B, Rosales-Mendoza S. Current sta[71] Salazar-González JA, Banuelos-Hernández tus of viral expression systems in plants and perspectives for oral vaccines development. Plant Mol Biol 2015;87:203–17. [72] García-Hernández AL, Rubio-Infante N, Moreno-Fierros L. Mucosal immunology and oral vaccination. In: Genetically engineered plants as a source of vaccines against wide spread diseases diseases—an integrated view. New York, NY: Springer; 2014. p. 15–42. [73] Gleba Y, Klimyuk V, Marillonnet S. Magnifection—a new platform for expressing recombinant vaccines in plants. Vaccine 2005;23:2042–8. [74] Granell A, Fernández del-Carmen A, Orzáez D. In planta production of plant-derived and non-plant-derived adjuvants. Expert Rev Vaccines 2010;9:843–58. [75] Kwon KC, Verma D, Singh ND, Herzog R, Daniell H. Oral delivery of human biopharmaceuticals, autoantigens and vaccine antigens bioencapsulated in plant cells. Adv Drug Delivery Rev 2013;Jun (65):782–99. [76] Scotti N, Rybicki EP. Virus-like particles produced in plants as potential vaccines. Expert Rev Vaccines 2013;12:211–24. [77] Kim MY, Reljic R, Kilbourne J, Ceballos-Olvera I, Yang MS, ReyesDel Valle J, et al. Novel vaccination approach for dengue infection based on recombinant immune complex universal platform. Vaccine 2015, http://dx.doi.org/10.1016/j.vaccine.2015.02.036 (pii: S0264410X(15)00225-X). [78] Langridge W, Dénes B, Fodor I. Cholera toxin B subunit modulation of mucosal vaccines for infectious and autoimmune diseases. Curr Opin Investig Drugs 2010;11:919–28. [79] Kohli N, Westerveld DR, Ayache AC, Verma A, Shil P, Prasad T, et al. Oral delivery of bioencapsulated proteins across blood–brain and blood–retinal barriers. Mol Ther 2014;22:535–46. [80] Daniell H, Singh ND, Mason H, Streatfield SJ. Plant-made vaccine antigens and biopharmaceuticals. Trends Plant Sci 2009;14:669–79. [81] Tacket CO, Mason HS, Losonsky G, Clements JD, Levine MM, Arntzen CJ. Immunogenicity in humans of a recombinant bacterial antigen delivered in a transgenic potato. Nat Med 1998;4:607–9. [82] Gomord V, Fitchette AC, Menu-Bouaouiche L, Saint-Jore-Dupas C, Plasson C, Michaud D, et al. Plant-specific glycosylation patterns in the context of therapeutic protein production. Plant Biotechnol J 2010;8:564–87. [83] Strasser R, Stadlmann J, Svoboda B, Altmann F, Glossl J, Mach L. Molecular basis of N-acetylglucosaminyltransferase I deficiency in Arabidopsis thaliana plants lacking complex N-glycans. Biochem J 2005;387:385–91. [84] Schähs M, Strasser R, Stadlmann J, Kunert R, Rademacher T, Steinkellner H. Production of a monoclonalantibody in plants with a humanized Nglycosylation pattern. Plant Biotechnol J 2007;5:657–63. [85] Gomord V, Faye L. Posttranslational modification of therapeutic proteins in plants. Curr Opin Plant Biol 2004;7:171–81. [86] Laissue J, Cottier H, Hess MW, Stoner RD. Early and enhanced germinal center formation and antibody responses in mice after primary stimulation with antigen-isologous antibody complexes as compared with antigen alone. J Immunol 1971;107:822–31. [87] Nie X, Basu S, Cerny J. Immunization with immune complex alters the repertoire of antigen-reactive B cells in the germinal centers. Eur J Immunol 1997;27:3517–25. [88] Regnault A, Lankar D, Lacabanne V, Rodriguez A, Thery C, et al. Fc gamma receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization. J Exp Med 1999;189:371–80. [89] Schuurhuis DH, Ioan-Facsinay A, Nagelkerken B, van Schip JJ, Sedlik C, et al. Antigen-antibody immune complexes empower dendritic cells to efficiently prime specific CD8+ CTL responses in vivo. J Immunol 2002;168: 2240–6. [90] Heyman B. The immune complex: possible ways of regulating the antibody response. Immunol Today 1990;11:310–3.
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840
G Model JVAC 16556 1–9
ARTICLE IN PRESS J.A. Salazar-González et al. / Vaccine xxx (2015) xxx–xxx
841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 Q6 859 860 861 862 863 864 865 Q7 866 867 868 869 870 871 872 873 874 875
[91] Chargelegue D, Drake PM, Obregon P, Prada A, Fairweather N, et al. Highly immunogenic and protective recombinant vaccine candidate expressed in transgenic plants. Infect Immun 2005;73:5915–22. [92] Pepponi I, Diogo GR, Stylianou E, van Dolleweerd CJ, Drake PMW, Paul MJ, et al. Plant-derived recombinant immune complexes as selfadjuvanting TB immunogens for mucosal boosting of BCG. Plant Biotechnol J 2014;12:840–50. [93] Phoolcharoen W, Bhoo SH, Lai H, Ma J, Arntzen CJ, et al. Expression of an immunogenic Ebola immune complex in Nicotiana benthamiana. Plant Biotechnol J 2011;9:807–16. [94] Cunha T, Aires-Barros R. Large-scale extraction of proteins. Mol Biotechnol 2002;20:29–40. [95] Floss DM, Schallau K, Rose-John S, Conrad U, Scheller J. Elastin-like polypeptides revolutionize protein expression and their biomedical application. Trends Biotechnol 2010;28:37–45. [96] Floss DM, Mockey M, Zanello G, Brosson D, Diogon M, Frutos R, et al. Expression and immunogenicity of the mycobacterial Ag85B/ESAT-6 antigens produced in transgenic plants by elastin-like peptide fusion strategy. Biomed Biotechnol 2010:2010. [97] Phan HT, Pohl J, Floss DM, Rabenstein F, Veits J, Le BT, et al. ELPylated haemagglutinins produced in tobacco plants induce potentially neutralizing antibodies against H5N1 viruses in mice. Plant Biotechnol J 2013;11:582–93. [98] Rubio-Infante N, Govea-Alonso DO, Romero-Maldonado A, García-Hernández AL, Ilhuicatzi-Alvarado D, Salazar-González JA, et al. A plant-derived multiHIV antigen induces broad immune responses in orally immunized mice. Mol Biotechnol 2015. [99] Govea-Alonso DO, Rubio-Infante N, García-Hernández AL, Varona-Santos JT, Korban SS, Moreno-Fierros L, et al. Immunogenic properties of a lettucederived C4(V3)6 multiepitopic HIV protein. Planta 2013;238:785–92. ˜ B, Hernández M, Fragoso G, Garate [100] Monreal-Escalante E, Banuelos-Hernández T, Sciutto E, et al. Expression of multiple Taenia solium immunogens in plant cells through a ribosomal skip mechanism. Mol Biotechnol 2015. [101] Yu J, Langridge WH. A plant-based multicomponent vaccine protects mice from enteric diseases. Nat Biotechnol 2001;19:548–52. [102] Lamichhane A, Azegamia T, Kiyonoa H. The mucosal immune system for vaccine development. Vaccine 2014;32(Nov (49)):6711–23.
9
[103] Rosales-Mendoza S, Salazar-González JA. Immunological aspects of using plant cells as delivery vehicles for oral vaccines. Expert Rev Vaccines 2014;13:737–49. [104] Pniewski T. Is an oral plant-based vaccine against hepatitis B virus possible. Curr Pharm Biotechnol 2012;13:2692–704. [105] Shamloul M, Trusa J, Mett V, Yusibov V. Optimization and utilization of Agrobacterium-mediated transient protein production in Nicotiana. J Vis Exp Q8 2014;19(Apr (86)). [106] Wirz H, Sauer-Budge AF, Briggs J, Sharpe A, Shu S, Sharon A. Automated production of plant-based vaccines and pharmaceuticals. J Lab Autom 2012;17(Dec (6)):449–57. [107] Yusibov V, Kushnir N, Streatfield SJ. Advances and challenges in the development and production of effective plant-based influenza vaccines. Expert Rev Vaccines 2015;14(4):519–35. [108] Shoji Y, Prokhnevsky A, Leffet B, Vetter N, Tottey S, Satinover S, et al. Immunogenicity of H1N1 influenza virus-like particles produced in Nicotiana benthamiana. Hum Vaccin Immunother 2015;11:118–23. [109] Landry N, Pillet S, Favre D, Poulin JF, Trépanier S, Yassine-Diab B, et al. Influenza virus-like particle vaccines made in Nicotiana benthamiana elicit durable, poly-functional and cross-reactive T cell responses to influenza HA antigens. Clin Immunol 2014;154:164–77. [110] Finer JJ. Plant nuclear transformation. In: Kempken F, Jung C, editors. Genetic modification of plants, biotechnology in agriculture and forestry, vol. 64. Berlin Heidelberg: Springer-Verlag; 2010. p. 3–21. [111] Sheludko YV. Agrobacterium-mediated transient expression as an approach to production of recombinant proteins in plants. Recent Pat Biotechnol 2008;2(3):198–208. [112] Verma D, Daniell H. Chloroplast vector systems for biotechnology applications. Plant Physiol 2007;145:1129–43. [113] Matoba N, Kajiura H, Cherni I, Doran JD, Bomsel M, Fujiyama K, et al. Biochemical and immunological characterization of the plant-derived candidate human immunodeficiency virus type 1 mucosal vaccine CTB-MPR. Plant Biotechnol J 2009;7:129–45. [114] Rybicki EP. Plant-based vaccines against viruses. Virol J 2014;11:205.
Please cite this article in press as: Salazar-González JA, et al. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.104
876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909