You are on page 1of 13
6122123, 12:05 AM "New human rabies vaccines in the pipeline - PMC Vaccine. 2019 Oct 3; 37(Suppl 1): A140-A145. PMCID: PMc6863069 dok: 10.1016)vaccine.2018.08.039: 10.1016/j.vaccine.2018.08.039 PMID: 30153997 New human rabies vaccines in the pipeline Anthony R. Fooks,* Ashley C. Banyard.* and Hildegund C.J, Ext "Animal and Plant Health Agency, Weybridge, Surrey, UK *wistar Institute, Philadelphia, PA, USA ‘Anthony R. Fooks: Tony.Fooks@apha.gov.uk; Ashley C. Banyard: Ashley Banyard@apha.gowuk; Hildegund C.J. Ent: ert@wistarorg “Corresponding author. ertl@wistar.org int © 2018 Published by Elsevier Ltd This is an open access article under the CC BY license (hitp:/Icreativecommons orgilicenses/by/3.0/), Abstract Rabies remains endemic in more than 150 countries. In 99% of human cases, rabies virus is transmitted by dogs. The disease, which is nearly always fatal, is preventable by vaccines given either before and/or after exposure to a rabid animal. Numerous factors including the high cost of vaccines, the relative complexity of post-exposure vaccination protocols requiring mul- tiple doses of vaccine, which in cases of severe exposure have to be combined with a rabies im- mune globulin, lack of access to health care, and insufficient surveillance contribute to the esti- mated 59,000 human deaths caused by rabies each year. New, less expensive and more im- munogenic rabies vaccines are needed together with improved surveillance and dog rabies control to reduce the death toll of human rabies. Here, we discuss new rabies vaccines that are in clinical and pre-clinical testing and evaluate their potential to replace current vaccines. Keywords: Pre-exposure prophylaxis (or PrEP), Post-exposure prophylaxis (PEP), Rabies immune globulin (RIG), Correlates of protection 1. Introduction Although vaccines that can prevent a rabies virus infection are commercially available, the death rate due to rabies encephalitis has more or less remained steady for decades. Human ra- bies occurs in 150 countries around the globe causing about 59,000 human deaths each year; although due to underreporting from developing countries the actual number is thought to be far higher [1]. Most cases are reported from Africa and Asia and ~40% of deaths affect chil- dren under the age of 14, Rabies is most commonly (more than 99% of cases) transmitted by dogs and although other mammals also transmit rabies virus; they account for less than 1% of human rabies cases. tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable a3 ovz2raa, 205m Now human rabies vaccines inthe pipeline - PMC Rabies virus is a member of the Lyssavirus genus. Lyssaviruses, all 16 of which are capable of causing rabies, are generally divided into two major phylogroups. Phylogroup I includes Rabies lyssavirus, Aravan lyssavirus, Australian bat lyssavirus, Bokeloh lyssavirus, Duvenhage lyssavirus, European bat-1 and bat-2 lyssaviruses, Irkut lyssavirus, Khujand lyssavirus, and Gannoruwa bat lyssavirus, while Lagos bat lyssavirus, Mokola lyssavirus, and Shimoni bat lyssavirus belong to phylogroup II [2]. Three further lyssaviruses, Ikoma lyssavirus, West Caucasian bat virus, and Lleida bat lyssavirus have also been characterized that fall outside of these two defined phylogroups. The majority of these viruses have been identified in bats and bats have been proposed as the host reservoir for all lyssaviruses with only Rabies lyssavirus commonly circulating in terrestrial carnivores. Existing vaccines are based on Rabies lyssavirus and protect against phylogroup | lyssaviruses but limited antigenic cross-reactivity between phylogroup I and II viruses precludes reliable protection against the latter (3], [4] Rabies, once symptomatic, is fatal in 99.9% of human cases and most of the human survivors have retained significant neurological sequalae, Rabies virus once transmitted by the saliva of an infected animal or through contact with infected tissues enters peripheral nerve endings and through axonal transport reaches the central nervous system (CNS). In the CNS, the virus replicates and then travels back to highly innervated tissues such as salivary glands from where it can be transmitted to the next victim. The incubation time for human rabies is highly variable. On average initial symptoms such as discomfort at the infection site, fever or general weakness commence 3-6 weeks after exposure [5] and then rapidly progress to a full-blown infection that can either take a furious or paralytic form. Patients with furious rabies exhibit symptoms of anxiety, confusion, fear of water due to problems swallowing, agitations and hal- lucinations while paralytic rabies patients progress from an ascending motor weakness to a coma [6]. In approximmately 1-3% of rabies cases the incubation time exceeds 6 months and can even extend over several years [7], [8]- ‘Treatment of symptomatic rabies has been attempted and the so-called Milwaukee protocol [9], which involves intensive therapy during which patients are shielded from neurological stimulations by coma-inducing drugs, and has reported limited success in a few patients. In most patients this protocol [10] and other experimental treatments such as anti-viral drugs, high doses of interferons [11], [12] or transfer of neutralizing antibodies [13] may prolong disease but do not avert death. Disease can be prevented by vaccines. Rabies vaccines have been available for over a century since the pioneering work of Pasteur and Roux in 1885 established that immunization with desiccated nerve tissue from an infected rabbit was able to prevent disease in a 9-year old boy, who had been attacked by a rabid dog. Since this time, safe and efficacious rabies vaccines have been developed. Despite this, thousands of humans are estimated to still die each year from this dreaded disease with the reasons behind this dichotomy being largely socioeconomic [14], The majority of humans at risk do not receive PEP after contact with a rabid animal or they receive PEP without RIG, Rabies vaccines and RIG are expensive, and the former have to be given repeatedly, Patients or their parents not only have to carry the costs for the biologi- cals, medical supplies and treatment but also encounter expenses due to repeated travel to health care facilities and loss of wages. Lack of knowledge of the disease and potential post ex- posure actions, lack of surveillance in animals and social instability further contribute to ab- sence of essential treatment tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable 23 12023, 12.05 aM Now human rabies vaccines inthe pipeline - PMC In individuals at high risk for rabies virus exposure the vaccines, which are based on inacti- vated purified viruses, are given in a pre-immunization (PrEP) regimen, Rabies vaccines are not overly immunogenic compared to live attenuated vaccines and contain no adjuvant so that 3 doses given are needed to reliably induce protective titers of neutralizing antibodies. Different regimens have been approved by the World Health Organization (WHO) [15] such as one-week (days 0, 3 and 7) two-site intradermal, two-week, one-site intramuscular (days 0, 3, 7 and between days 14-28) and 3 week-intramuscular (days 0 [2 sites], 7 [one site] and 21 [1 site] vaccinations, Immunological memory is long-lasting [16], [17] so that upon rabies virus exposure of an already vaccinated individual, one or two doses of vaccine are administered that according to WHO should be given at one site intradermally on days 0 and 3, four sites in- tradermally only on day 0 or one site intramuscularly on days 0 and 3 [15]. In an unvaccinated individual upon contact with a rabid animal, the severity of exposure dictates if the vaccine has to be combined with a rabies immune serum (RIG). In cases of licks or minor scratches, three regimens (5-and 4-dose regimen, 2-1-1 regimen) are approved for intramuscular post-expo- sure prophylaxis (PEP) and a 2 site 4-dose intradermal regimen has been adapted by some countries. In cases of more invasive exposures, such as bites, licks of mucosal membranes or exposure to bats, the vaccine needs to be combined with RIG; and most of the RIG should be infiltrated into the wound to prevent further spread of the virus. In either case wounds need to be cleaned thoroughly to neutralize as many live virions as possible [15] 2. Correlates of protection Correlates of protection {18], [19] against rabies virus are well defined. Neutralizing antibod- ies directed against the rabies virus glycoprotein [20], the only surface protein of this bullet- shaped virus, prevent disease. In general titers of or above 0.5 international units/ml are evi- dence of seroconversion and considered protective [21] but this number should be viewed with caution [22]. Rabies causing lyssaviruses are a diverse group of viruses. Higher titers, which are typically determined by tests based on Rabies virus, may be needed to reliably pro- tect against lyssaviruses that are antigenically more distant from the vaccine strain. Induction of an immunoglobulin class switched affinity-matured B cell response [23], [24] requires help from CD4* T cells, Although this has not yet been studied for rabies vaccines, B cell help is pro- vided best by follicular T help cells, which in germinal centers within lymphatic tissues provide important cues for differentiation of naive B cells into long-lived antibody secreting cells and memory B cells [25]. Individuals with inherited or acquired B or CD4" T cell immunodeficien- cies will not respond well to a rabies vaccine [26], [27]. In these patients, the use of RIG is mandatory after exposure, and titers should be checked after vaccination and if they remain below 0.5 IU/ml additional doses of vaccine should be given [28]. As testing of antibody titers is costly, this protocol is rarely followed in resource poor-countries. 3. Current rabies vaccines Early rabies vaccines were modified versions of the vaccine that was used in 1885 by Louis Pasteur. They were produced in nerve tissue and then inactivated with phenol. For PEP, 14-21 daily doses were needed to induce adequate titers of antibodies. The potency of these so- called Semple vaccines was highly variable. As Semple vaccines contained residual nerve tissue neurological complications such as neuropathy, Guillain-Barré syndrome, meningitis or en- cephalitis were common, Nerve tissue rabies vaccines (NTVs) including the Semple and the Fuenzalida rabies vaccine, the latter being grown in suckling mouse brain and thus containing hitps:tnwwznebilm nih. govlpmelaticies!PMC5863068/?report=printable ans 12023, 12.05 aM Now human rabies vaccines inthe pipeline - PMC less myelin, are still used in some developing countries [29]. However, NTVs are no longer rec- ommended by the WHO, which endorses the use of vaccines grown in cell culture or embry- onated eggs. For WHO approved vaccines rabies virus grown in embryonated duck or chicken eggs, primary chick embryo cells, Vero cells or human diploid cells is upon harvest concen trated, purified, inactivated and lyophilized. Potency is tested, and the vaccine is required to contain equal or above 2.5 international units per intramuscular dose [14]. 4. Future human rabies vaccines Next generation human vaccines are in general developed to reduce adverse events or im- prove efficacy of available vaccines. While early nerve tissue rabies vaccines were highly reac- togenic, current vaccines have an excellent safety profile and severe adverse events are scarce, Available rabies vaccines are highly efficacious and upon their appropriate use vaccine failures have been reported [30] but are very rare. Economic factors, rather than concerns about safety or lack of efficacy, are driving the development of new rabies vaccines. Reduced cost and, preferentially, single immunization regimens would be especially valuable to expand pre- exposure prophylaxis (PrEP) and allow for its incorporation into childhood immunization pro- grams in highly endemic areas. The effectiveness of more wide-spread PrEP has been validated in Peru [31]. This country has successfully stopped the increasing incidence of vampire bat- transmitted rabies in children in Amazonia by incorporating the rabies vaccine into routine childhood immunization programs (31). Nevertheless, the use of current rabies vaccines for childhood PrEP is not cost-effective [32], which necessitates the development of cheaper and ‘more immunogenic single dose vaccines that are stable and do not require cold-chain storage and can be preferentially administered orally or needle-free [33] The same rabies vaccines are used for both PrEP and PEP. PrEP aims to induce sustained titers of rabies virus neutralizing antibodies and immunological memory. The goal of PEP is to stimu- late a rapid neutralizing antibody response following potential exposure. The initial short-lived antibody response composed of IgM develops outside germinal centers and is independent of T cell help. Neutralizing IgM antibodies appear within a few days and, as studies in mice have shown, may contribute to protection [34]. Induction of sustained affinity-matured IgG neutral- izing antibody responses and memory B cells takes longer but is essential for PrEP. One could thus argue that different vaccines should be developed to fulfill the needs of PrEP versus PEP. Subunit vaccines based on genetic sequences that encode the rabies virus glycoprotein can de- pend on the delivery vehicle to induce a potent and sustained antibody and memory B cell re- sponses after a single dose [35]. It takes time for genetic vaccines to first enter cells, become transcribed and translated and produce enough protein to induce an immune response. Genetic vaccines although highly suited for PrEP should thus not be developed for PEP. Adjuvanted rabies vaccines, vaccines based on genetically modified rabies virus or protein vac- cines on the other hand are appropriate for PEP. 5. Protein vaccines The rabies virus glycoprotein assembles as a homotrimeric protein on the surface of the virion and most of the antigenic sites are conformational epitopes that depend on the correct quater- nary protein structure [36]. Generating cost-effective protein-based vaccines with the correct structure remains a challenge, especially when a crystal-structure for lyssavirus glycoprotein remains undefined. However, a number of expression systems have been explored, Mammalian hitps:tnwwzncb.m nih. govlpmelaticies!PMC88653068/?report=printable ans. 12023, 12.05 aM Now human rabies vaccines inthe pipeline - PMC expression systems based on human embryonic kidney (HEK) 293 [37], baby hamster kidney (BHK)-21 [38], or Chinese hamster ovary (CHO) [39] cell lines have been tested with varied success; they showed distinct patterns of glycosylation depending on cell substrate and culture conditions [40]. Insect cell expression system based on recombinant baculovirus [41] or trans- fected Drosophila melanogaster Schneider 2 cells [42] tend to be more efficient for high yield protein expression but they typically add shorter glycans with added fucose and xylose residues. Yeast expression system are also cost-effective by their glycosylation with mannose only containing glycans is very different from that of mammalian cells and accordingly yeast- derived rabies virus glycoprotein is poorly immunogenic [43]. The rabies virus glycoprotein has also been expressed by plants such as tomatoes [44], carrots [45], maize [46] and others with the intention to eventually develop edible rabies vaccines, Producing foreign proteins in plants is cost-effective but hampered by purification issues. The latter would not affect edible vaccines but immune responses elicited by oral immunization with plant-derived rabies virus glycoprotein have thus far been variable and in general below those needed to achieve reliable protection [47]. One protein nanoparticle vaccine generated by CPI. Biologicals, Gujarat, India, is currently in phase Ill testing in healthy volunteers in India (https://biospectrumasia.com/article/pdf/9246). This vaccine is based on a recombinant bac- ulovirus-derived glycoprotein that was modified to self-assemble into nanoparticles. Information on the performance of the vaccine in Phase I/II trials is not available. Although protein vaccines are safe they may not be cost effective. They would require extensive purifica- tion and addition of an adjuvant. Experiments in pre-clinical models showed that they would not be sufficiently immunogenic to allow for a 1-dose regimen. 6. Adjuvanted rabies vaccines Adjuvants enhance immune responses through so-called danger signals that drive maturation of immature dendritic cells into professional antigen presenting cells or by providing antigen depots that prolong antigen presentation, Alum, the most commonly used adjuvant, does not increase immune responses to the rabies vaccine [48]. Second generation adjuvants based on ligands for Toll-like receptors (TLR) seem to perform better [49]. One adjuvanted rabies vac- cine called PIKA vaccine composed of Rabipur and a polyinosinic-polycytidylic acid based, TLR- 3-activating adjuvant, has completed a phase II clinical trial [50]. ‘The vaccine was given in an accelerated regimen with 2 doses on day 0, 2 doses on day 3 and one dose on day 7 and com- pared to Rabipur given at the regular day 0, 3, 7 and 14 regimen. The PIKA vaccine was well tolerated, Antibody titers 20.5 IU/ml increased faster than with the non-adjuvanted vaccine. Overall the trial showed comparative efficacy of the PIKA vaccine to the currently used rabies vaccine, Larger scale trials are needed to assess if the adjuvanted vaccine reliably accelerates onset of protective titers of rabies virus neutralizing antibodies, which would be highly advan- tageous for PEP. Other adjuvants have been tested pre-clinically in combination with the rabies vaccine and in general results indicated that they allowed for dose-sparing [51], [52], [53]. 7. Genetically modified rabies virus Rabies virus can be modified by genetic engineering [54]. Genes such as those encoding the phosphoprotein [55] or the matrix protein [56] can be deleted which attenuates the virus. Rabies viruses with such deletions do not replicate in animals including those with immunode- ficiencies and are thus presumably safe, Matrix protein-deleted rabies virus is more immuno- genic than virus with a phosphoprotein deletion or inactivated wild-type virus [57]. In spite of hitps:tnwwznebi.alm nih. govipmelaticies!PMC58653068/?repert=printable 53 12023, 12.05 aM Now human rabies vaccines inthe pipeline - PMC such promising immunogenicity data, the modified viruses grow less vigorously in cell culture than wild-type virus, which will likely raise production issues and increase vaccine cost. Also, public acceptance of a live rabies vaccine might be limited. Alongside using deletion mutants, vaccine viruses carrying two or three glycoprotein gene copies have also been developed [58]. In murine studies, inactivated virus showed increased immunogenicity compared to wild-type virus. Such viruses, provided their growth allows for scaled production, could be explored for PrEP. 8. Genetic vaccines Genetic vaccines can be divided into RNA vaccines, DNA vaccines and viral vector vaccines. ‘They have in common that a gene rather than a protein antigen is transferred. In case of RNA or DNA vaccines the gene only encodes for a rabies virus antigen, which results in a highly fo- cused immune response unlike viral vectors, which induce T and B cell responses to antigens of the gene carrier as well. Uptake of different types of genetic vaccines depends on the vac- cine carrier. Viral vectors commonly bind to specific cell surface receptors and are therefore readily internalized, Nevertheless, pre-existing neutralizing antibodies to viral vectors induced by previous natural infections can severely dampen their uptake and immune responses to a transgene product [59], [60]. Similarly, a viral vector should not be given repeatedly as vector neutralizing antibodies induced by the first immunization will impact the effectiveness of sub- sequent boosts. This is not a concern for RNA or DNA vaccines. Transfer of RNA or DNA vac- cines across cell membranes is rather inefficient but can be facilitated by various physical or chemical methods [61] 9. RNA vaccines A clinical phase | trial with an RNA rabies vaccine expressing the rabies virus glycoprotein, termed CV7201, tested a three-dose regimen in healthy adult volunteers. The vaccine was well tolerated but protective titers of antibodies were not achieved in all vaccinated individuals, titers declined by one year after vaccination and upon an additional boost failed to increase in all participants [62]. The RNA vaccine was thus clearly inferior to commercially available vaccines, 10. DNA vaccines Multiple pre-clinical trials have shown that DNA vaccines given to mice [63], [64J, [65] or mon- keys [66], [67], [68] protect against rabies virus in PrEP or even PEP regimens. DNA vaccines against diseases other than rabies have been tested in multiple phase | and II trials with varied success, On phase IIb studies that tested a prime boost regimen composed of a DNA vaccine prime followed by an adenovirus vector boost for prevention of HIV-1 infection failed to meet its endpoint, and was stopped prematurely [69]. Another phase III trial for a CMV vaccine by Astellas also failed (Clinical Trials gov Identifier: NCT01877655). An additional phase Il trial following promising phase II results [70] has been initiated with a therapeutic DNA vaccine for oncogenic types of human papillomavirus-associated tumors. The vaccine is delivered by elec- troporation and it should be pointed out that it aims to induce T cell responses rather than a rabies DNA vaccine that needs to target B cells, tps stww.ncb.nim.nih.govlpmetarticles/MC6863069)report=pintable ens. 12023, 12.05 aM New human rables vaccines in the pipeline - PMC Thus far overall available clinical data show that DNA vaccines are poorly immunogenic in hu- mans. This may in part be overcome by improved delivery methods such as electroporation, Whether the use of such devices is practical in developing countries remains to be explored. 11. Viral vector vaccines The safest and most immunogenic of the different types of viral vector vaccines are based on El-deleted adenoviruses [71]. Adenovirus vectors are safe for the E1 deletion renders them replication-defective. There are several potential reasons why they are exceptionally immuno- genic compared to other viral vectors. The E1-deletion reduces transcription of the viral anti- gens without affecting expression of the transgene product, which is typically under the con- trol of a potent ubiquitously active promoter. This focuses the immune response largely on the transgene product rather than on antigens of the vaccine carrier. Adenovirus vectors are not cytolytic and they persist at low levels in a transcriptionally active form for an extended period of time thus constantly providing internal boost to the immune system. Immune responses to e [72]. Non-human serotypes have been vectored [73], which avoids potential dampening of vaccine efficacy by pre-existing neutralizing antibodies to the vaccine carrier [74]. In non-human primates adenovirus vectors expressing the rabies virus glycoprotein were shown to induce long-lasting protective titers of neutralizing antibodies after a single dose [35], [75]. As adenoviruses persist albeit a low doses in a transcriptionally active form antibody titers are sustained [72]. Methods for storage without need for a cold-chain have been developed facilitating the use of such vaccines in re- source-poor countries [76]. Ithas been estimated that upon mass production the cost of a sin- gle dose adenovirus rabies vaccine would be significantly lower than that of a 3-dose course of a traditional rabies vaccines [77]. This in turn would allow for their use in childhood immuni- zation programs in highly endemic regions. adenovirus vectors thus remain stable over long periods of Other recombinant viral vectors have been explored. They were either too reactogenic, lacked efficacy or required prime-boost regimens. Furthermore, most them have not yet undergone clinical testing as vaccine carriers and it is thus unclear if methods for scale-up, purification and release testing that meet regulatory requirements can be achieved. Poxvirus vectors have been tested extensively in experimental animals [78], [79], [80], [81]. They are less immuno- genic than adenoviral vectors [82], which may in part relate to a dampening of immune re- sponses to the transgene product by potent immune responses to the numerous antigens of the vaccine carrier. Poxvirus vectors based on vaccinia virus are also reactogenic [83], while those based on more attenuated poxviruses such as Modified Vaccinia Ankara lack efficacy [81]. A rabies vaccine based on parainfluenza virus showed efficacy in mice if given twice in a prime-boost regimen; single dose regimens were not explored [84]. Long-lasting protection of dogs was observed upon a single immunization with a high dose of a Newcastle Disease Virus (NDV) rabies vaccine [85]. NDV is highly pathogenic for birds and itis transmissible to hu- mans; potential toxicity of a recombinant NDV in either species needs to be explored further. A single dose of an attenuated pseudorabies virus expressing the rabies virus glycoprotein in- duced neutralizing antibodies in dogs [86]. The study did not assess protection against challenge. tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable 73 6122123, 12:05 AM "New human rabies vaccines in the pipeline - PMC 12. Summary Next generation human rabies vaccines are needed to reduce cost and numbers of doses for PrP and PEP, It could be advantageous to develop different vaccine platforms for these two applications as each has their unique requirement for optimally protective immune responses. For PrP vaccines that induce long-lived rabies virus neutralizing antibody and sustained memory B cell responses after a single dose would be needed and replication-defective aden- oviral vectors may fulfill these requirements. There is currently no human rabies vaccine plat- form on the horizon that has been shown pre-clinically in a relevant animal model such as non-human primates to accelerate antibody production upon PEP and allow for a reduction in vaccine doses and potentially RIG. Further tools are stil required to prevent rabies replication following the onset of clinical disease. For review of rabies virus, epidemiology, diagnosis, prevention, and management: consult Fooks et al. in Nature Reviews Disease Primers [87]. Conflict of Interest Statement None declared References 1, Hampson K,, Coudeville L., Lembo T,, Sambo M., Kieffer A., Attlan M, Estimating the global burden of endemic canine rabies. PLoS Neg! Trop Dis. 2015;9 [PMCID: PMC4427501] [PubMed: 25961848] 2, Amarasinghe G.K,, Aréchiga Ceballos N.G,, Banyard A.C, Basler C.R, Bavari S, Bennett A, Taxonomy of the order mononegavirales: update 2018, Arch Virol. 2018 [PMCID: PMC6076851) [PubMed: 29637429] 3. Brookes SM, Healy D.M., Fooks AR. Ability of rabies vaccine strains to elicit cross-neutralising antibodies. Dev Biol (Basel) 2006;125:185-193. [PubMed: 16878476] 4. Badrane HL, Bahloul C, Perrin P, Tordo N. Evidence of two Lyssavirus phylogroups with distinct pathogenicity and immunogenicity, J Virol, 2001;75:3268-3276, [PMCID: PMC114120] [PubMed: 11238853] 5, Plotkin S.A. Rabies. Clin infect Dis, 2000;30:4-12. (PubMed: 10619725] 6, Warrell D.A. The clinical picture of rabies in man. Trans R Soc Trop Med Hyg. 1976;70:188-195, [PubMed: 982512] 7. Boland T.A., McGuone D,, Jindal J, Rocha M., Cumming M., Ruprecht CE. Phylogenetic and epidemiologic evidence of multiyear incubation in human rabies, Ann Neurol, 2014;75:155-160, [PMCID: PMC4118733] [PubMed: 24038455] 8, Shankar S.K., Mahadevan A,, Sapico S.D., Ghodkirekar M.S.G, Pinto R.G.W,, Madhusudana S.N. Rabies viral ‘encephalitis with proable 25 year incubation period! Ann Indian Acad Neurol. 2012;15:221-223, [PMCID: PMC3424805] [PubMed: 22919200] 9, Willoughby RE, Tieves KS, Hoffman G.M., Ghanayem N.S, Amlie-Lefond CM, Schwabe M.J, Survival after treatment of rabies with induction of coma, N Engl Med. 2005;352:2508-2514. [PubMed: 15958806] 10. Zeiler FA, Jackson A.C. Critical appraisal of the Milvwaukee protocol for rabies: this failed approach should be abandoned, Can J Neurol Sct, 2016;43:44-51. [PubMed: 26639059] tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable ans 6122123, 12:05 AM "New human rabies vaccines ir he pipeline - PMC 11, Warrell M,., White N,J., Looareesuwan S, Phillips R.E, Suntharasamai P, Chanthavanich P, Failure of interferon alfa and tribavirin in rabies encephalitis. BM). 1989;299:830-833, [PMCID: PMC1837695] [PubMed: 2510846] 12, Merigan T.C, Baer G.M., Winkler W.G,, Bernard K.W,, Gibert CG, Chany C. Human leukocyte interferon administration to patients with symptomatic and suspected rabies, Ann Neurol. 1984;16:82-87, [PubMed: 6205625] 13. Hemachudha T, Sunsaneewitayakul B., Mitrabhakdi E., Suankratay C, Laothamathas J, Wacharapluesadee S. Paralytic complications following intravenous rabies immune globulin treatment in a patient with furious rabies. Int] Infect Dis, 2003;7:76-77. [PubMed: 12718814] 14. Wilde H., Tipkong P, Khawplod P Economic issues in postexposure rabies treatment. Travel Med. 1999;6:238-242. [PubMed 10575172] 15, World Health Organization, WHO Expert Consultation on Rabies. Third report. World Health Organization Technical Report Series; 2018. p. 1-195 16. Thraenhart 0,, Kreuzfelder E, Hillebrandt M., Marcus 1, Ramakrishnan K,, Fu F Long-term humoral and cellular immunity after vaccination with cell culture rabies vaccines in man, Clin Immunol Immunopathol. 1994;71:287-292. [PubMed: 8200131) 17. Brown D., Featherstone JJ, Fooks AR, GettnerS,, Lloyd E,, Schweiger M. Intradermal pre-exposure rabies vaccine clicits long lasting immunity. Vaccine. 2008;26:3909-3912. [PubMed: 18547689] 18, Johnson N,, Cunningham AR, Fooks A.R, The immune response to rabies virus infection and vaccination, Vaccine. 2010;28:3896-3901. [PubMed: 20368119] 19, Horowitz A., Behrens R.H., Okell L,, Fooks A.R,, Riley EM, NK cells as effectors of acquired immune responses: effector CD4+ 1 cell-dependent activation of NK cells following vaccination, J Immunol. 2010;185:2808-2818, [PubMed: 20679529} 20, Xiang Z.Q,, Knowles B.B,, MeCarrick |.W, Ert! H.C. Immune effector mechanisms required for protection to rabies virus. Virology. 1995;214:398-404, [PubMed: 8553540] 21, WHO Expert Consultation on rabies. World Health Organ Tech Rep Ser. 2005; 931: 1-88, [PubMed: 16485446] 22. Malerczyk C, SelhorstT, Tordo N., Moore S,, Miller. Antibodies induced by vaccination with purified chick embryo cell culture vaccine (PCECV) cross-neutralize non-classical bat lyssavirus strains, Vaccine, 2009;27:5320- 5325, [PubMed: 19615958] 23, McKimmie CS, Johnson N,, Fooks A.R, Fazakerley J.X. Viruses selectively upregulate Toll-like receptors in the central nervous system. Biochem Biophys Res Commun, 2005;336:925-933. [PubMed: 16157304] 24, Mansfield KL, Johnson N., Nunez A., Hicks D., Jackson A.C, Fooks A.R. Up-regulation of chemokine gene transcripts and T-cell infiltration into the central nervous system and dorsal root ganglia are characteristics of experimental European bat lyssavirus type 2 infection of mice. J Newrovirol. 2008;14:218-228, (PubMed: 18569456] 25. Ma CS, Phan T.G. Here, there and everywhere: T follicular helper cells on the move. Immunology. 2017;152:382- 387, [PMCID: PMC5629422] [PubMed: 28704588] 26, Pancharven C, Thisyakorn U, Tantawichien T, Jaijaroensup W,, Khawplod P, Wilde H. Failure of pre- and postexposure rabies vaccinations in a child infected with HIV, Scand J Infect Dis. 2001;33:390-391. [PubMed: 11440231] 27. van Zelm M.C, Reisli L, van der Burg M., Castafo D., van Noesel C.M., van Tol M.D. An antibody-defictency syndrome due to mutations in the CD19 gene. N Engl Med, 2006;384:1901-1912. [PubMed: 16672701] tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable ans 6122123, 12:05 AM "New human rabies vaccines ir he pipeline - PMC 28, Hicks D.), Fooks A.R,, Johnson N, Developments in rabies vaccines, Clin Exp Immunol. 2012;169:199-204, [PMCID PMc3444995} [PubMed: 22861358] 29. Wu X, Smith T.G., Ruprecht CE. From brain passage to cell adaptation: the road of human rabies vaccine development, Expert Rev Vaccines. 2011;10:1597-1608, [PubMed: 22043958] 30, Wilde H. Failures of postexposure rabies prophylaxis. Vaccine. 2007;25:7605-7609. [PubMed 17905484] 31. Kessels |.A,, Recuenco S,, Navarro-Vela AM., Deray R., Vigilato M., Ertl H. Pre-exposure rabies prophylaxis: a systematic review. Bull World Health Organ. 2017;95:210-219C. (PMCID: PMC5328107] [PubMed: 28250534] 32, Chulasugandha P, Khawplod P, Havanond P, Wilde H. post-exposure treatment in Thai children, Vaccine, 2006;24:1478-1482, [PubMed: 16221511] Cost comparison of rabies pre-exposure vaccination with 33, Fooks A.R,, Koraka P, de Swart R.L., Rupprecht C.E., Osterhaus A.D.M.E, Development of a multivalent paediatric ‘human vaccine for rabies virus in combination with Measles-Mumps-Rubella (MMR) Vaccine. 2014;32:2020-2021. [PubMed: 24589425] 34, Dorfmeier CLL, Shen S,, Tzvetkov E,P, McGettigan |.P Reinvestigating the role of IgM in rabies virus postexposure vaccination. J Virol. 2013;87:9217-9222. [PMCID: PMC3754079] [PubMed: 23760250] 35. Xiang Z.Q., Greenberg L,, Ertl H.C., Ruprecht CE, Protection of non-human primates against rabies with an adenovirus recombinant vaccine, Virology. 2014;450~45 1:243-249, (PMCID: PMC4038128] | PubMed: 24503087] 36, Wunner W.H., Dietzschold B., MacFarlan Ril, Smith C.L,, Golub E,, Wiktor TJ. Localization of domains on the rabies virus glycoprotein. Annales de Institut Pasteur/Virologie. 1985;136:353-362, munogenic 37. Fontana D., Kratje R. 2014;32:2799-2804, (PubMed: 24631077] hheverrigaray M., Prieto C. Rabies virus-like particles expressed in HEK293 cells. Vaccine, 38. Ferndndex-Nilex E.G,, de Rezende AG. Puglia ALP, Leme }, Boldorini VIL rabies virus G-glycoprotein using BHK-21 cells cultured in suspension. Biotechnol Lett. 2015;37:1 153-1163. [PubMed 25700821] Caricati CP Transient expression of 39, Wojezyk B.S secreted form of recombinant rabies virus glycoprotein: comparison of two affinity tags. Protein Expr Purif: 1996;7:183-193, [PubMed: 8812859] Czerwinski M., Stwora-Wojezyk M.M., Siegel D.L., Abrams WR, Wunner WH. Purification of a 40. Brooks S.A. Appropriate glycosylation of recombinant proteins for human use: implications of choice of expression system, Mol Biotechnol, 2004;28:241-255, [PubMed: 15542924] 41. Prehaud C, Takehara K, Flamand A., Bishop D-H. Immunogente and protective properties of rabies virus slycoprotein expressed by baculovirus vectors. Virology. 1989;173:390-399. [PubMed: 2688298] 42, Galesi A.LiL,, Aguiar M.A., Astray RiM,, Augusto ‘melanogaster Schneider 2 cells producing rabies virus glycoprotein in bioreactor employing serum-free medium. (ytotechnology. 2008;57:73-81. [PMCID: PMC2553640] [PubMed: 19003175] P, Moraes A.M. Growth of recombinant Drosophila 43, Sakamoto S, Ide T, Tokiyoshi S, Nakao J, Hamada F, Yamamoto M. Studies on the structures and antigenic properties of rabies virus glycoprotein analogues produced in yeast cells. Vaccine. 1999;17-205~218. [PubMed: 9987156] 44, McGarvey PB, Hammond J., Dienelt M.M., Hooper D.C, Fu Z. glycoprotein in transgenic tomatoes. Biotechnology (NY) 1995;13:1484-1487. [PubMed: 9636308] Dietzschold B. Expression of the rabies virus 45, Rojas-Anaya E,, Loza-Rubio E,, Olivera-Flores M.., Gomez-Lim M, Expression of rabies virus G protein in carrots (Daucus carota) Transgenic Res, 2009;18:911-919, (PubMed: 19479338] tps stww.ncb.nim.nih.govlpmetarticles/MC6863069)report=pintable sons 6122123, 12:05 AM "New human rabies vaccines ir he pipeline - PMC 46, Loza-Rubio E,, Rojas F,, Gémez L,, Olivera M.T., Gémer-Lim M.A, Development of an edible rabies vaccine in maize using the Vaukovo strain, Dev Biol (Basel) 2008;131:477-482. [PubMed: 18634510] 47. Yusibov V,, Hooper D.C, Spitsin SV,, Fleysh N., Kean RB, Mikheeva T. Expression in plants and immunogenicity of plant virus-based experimental rabies vaccine, Vaccine. 2002;20:3155-3164, [PubMed: 12163267] 48. Yendo ACA, de Costa F, Cibulski SP, Teixeira TF, Colling L Mastrogiovanni M. A rabies vaccine adjuvanted with saponins from leaves of the soap tree (Quillaja brasiliensis) induces specific immune responses and protects against lethal challenge, Vaccine. 2016;34:2305-2311. [PubMed: 27032516] 49. Wang X, Bao M, Wan M., Wel H., Wang L., Yu H. A CpG oligodeoxynucleotide acts as a potent adjuvant for inactivated rabies virus vaccine. Vaccine. 2008;26:1893-1901. [PubMed: 18321616] 50. Kalimuddin S, Wijaya L., Chan YFZ., Wong AWLL, Oh HL.M.L., Wang L.-F A phase II randomized study to determine the safety and immunogenicity of the novel PIKA rabies vaccine containing the PIKA adjuvant using an accelerated regimen, Vaccine. 2017;35:7127-7132, (PubMed: 29174316] 51, Miao L., Yang ¥, Yan M., Li ¥, Zhao J, Guo J. Enhanced immune response to rabies viruses by the use of a liposome adjuvant in vaceines. Viral Immunol. 2017 [PubMed: 28953450] 52, Nicholson SM. A novel adjuvant promises improved rabies vaccine. Lab Anim (NY) 2016;45:128. [PubMed: 27003344] 53. Xiao X, Zhang ¥,, Liu, Wel Q., Yin X Immunoenhancement with flagellin as an adjuvant to whole-killed rabies vaccine in mice. Arch Virol. 2016;161:685-691. [PubMed: 26650039] 54, Mebatsion T, Schnell MJ, Cox JH, Finke S,, Conzelmann K.K, Highly stable expression of a foreign gene from rabies virus vectors. Proc Nat! Acad Sci USA. 1996;93:7310-7314. [PMCID: PMC38980] (PubMed: 8692989] 55, Ito Sugiyama M,, Yamada K,, Shimizu K,, Takayama-ito M., Hosokawa J, Characterization of M gene-deficient rabies virus with advantages of effective immunization and safety as a vaccine strain, Microbiol Immunol. 2005;49:971-979, [PubMed: 16301807] 56, Morimoto K,, Shoji ¥,, Inoue S, Characterization of P gene-deficient rabies virus: propagation, pathogenicity and antigenicity, Virus Res. 2005;111:61-67, [PubMed: 15896403] 57, Cenna,, Hunter M, Tan G.S,, Papaneri A.B., Ribka .P, Schnell MJ. Replication-deficient rabies virus-based vaccines are safe and immunogenic in mice and nonhuman primates. J Infect Dis. 2009;200:1251-1260, [PMCID: PMC3388800] [PubMed: 19764884) 58, Hosokawa-Muto J, Ito N., Yamada K., Shimizu K,, Sugiyama M,, Minamoto N. Characterization of recombinant rabies virus carrying double glycoprotein genes, Microbiol Immunol. 2006;50:187-196, [PubMed: 16547416] 59, Papp Z,, Babluk L.A., Baca-Estrada M.E, The effect of pre-existing adenovirus-specific immunity on immune responses induced by recombinant adenovirus expressing glycoprotein D of bovine herpesvirus type 1, Vaccine, 1999;17:933-943, [PubMed: 1006700] 660, Galletti R,, Beauverger P, Wild LR. Passively administered antibody suppresses the induction of measles virus measles recombinant viruses. Vaccine, 1995;13:197-201. [PubMed: 7625116] antibodies by vaccini: 61, Jorritsma S.H.T, Gowans B,J, Grubor-Bauk B,, Wijesundara DK, Delivery methods to increase cellular uptake and immunogenicity of DNA vaccines, Vaccine. 2016;34:5488-5494, [PubMed: 27742218] 62. Alberer M., Gnad-Vogt U, Hong ILS, Mehr K-T, Backert L., Finak G. Safety and immunogenicity of amRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first 2017;390:1511-1520, [PubMed: 28754494] 1uman phase 1 clinical trial. Lancet. tps stww.ncb.nim.nih.govlpmetarticles/MC6863069)report=pintable 3 6122123, 12:05 AM "New human rabies vaccines ir he pipeline - PMC 63, Xiang.Z.Q. Spitalnik SL, Cheng J, Erikson J Wojczyk B,, Ertl H.C, Immune responses to nucleic acid vaccines to '09:569-579, (PubMed: 7778289] rabies virus. Virology. 199: 64, Ray N.B,, EwaltL.C., Lodmell D.L. Nanogram quantities of plasmid DNA encoding the rabies virus glycoprotein protect mice against lethal rabies virus infection, Vaccine. 1997;15:892-895, [PubMed: 9234541] 65. Bahloul C, Jacob ¥, Tordo N., Perrin P DNA-based immunization for exploring the enlargement of immunological cross-reactivity against the lyssaviruses. Vaccine, 1998;16:417-425. [PubMed: 9607065] (66. Lodmell D.L., Parnell M.J, Bailey [.R, Ewalt L.C, Hanlon CA. Onestime gene gun or intramuscular rabies DNA vaccination of non-human primates: comparison of neutralizing antibody responses and protection against rabies virus 1 year after vaccination. Vaccine, 2001;20:838-844. (PubMed: 11738747] 67. Lodmell D.L., Parnell M.J,, Bailey [.R, Ewalt L.C, Hanlon GA. Rabies DNA vaccination of non-human primates: post- ‘exposure studies using gene gun methodology that accelerates induction of neutralizing antibody and enhances neutralizing antibody titers, Vaccine. 2002;20:2221-2228, [PubMed: 12009276] 68. Kumar B.D., Kumar PU, Krishna TP, Kalyanasundaram S, Suresh P, Jagadeesan V, Pre-clinical toxicity & immunobiological evaluation of DNA rabies vaccine & combination rabies vaccine in thesus monkeys (Macaca mulatta) Indian J Med Res. 2013;137:1072- 1088, [PMCID: PMC3734712] [PubMed: 23852288] 69, Hammer SM, Sobieszeay& MCE, Janes H, Karuna, Mulligan M.., Grove D. Efficacy trial ofa DNA/sAdS HV preventive vaccine, N Engl Med. 2013;269:2083-2092.[PMCID: PMC4030694][PubMed: 24099601] 70, Trimble C.L,, Morrow M.P, Kraynyak K.A, Shen X,, Dallas M., Yan J Safety, efficacy, and immunogenicity of VGX- 3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078- 2088, [PMCID: PMC4888059] [PubMed: 26386540] 71. Casimiro D.R,, Chen I., Fu T-M,, Evans RK, Caulfield MJ, Davies M.-E. Comparative immunogenicity in rhesus monkeys of DNA plasmid, recombinant vaccinia virus, and replication-defective adenovirus vectors expressing 2 ‘human immunodeficiency virus type 1 gag gene. J Virol 2003;77:6305-6313, [PMCID: PMC154996] [PubMed: 12743287] 72. Tatsis N., Fitegeral J.C. Reyes-Sandoval A, Harris-MeCoy K.C, Hensley SE, Zhou D. Adenoviral vectors persistin vivo and maintain activated CD8+T cells: implications for their use as vaccines, Blood. 2007;110:1916-1923. [PMCID: PMC1976365] [PubMed: 17510320] 73, Farina SF, Gao GP, Xiang7.Q., Rux J, Burnett R.M., Alvira M.R, Replication-defective vector based on a chimpanzee adenovirus, J Virol. 2001;75:11603-11613, [PMCID: PMC114747] [PubMed: 11689642] 74, Xiang Z., Li ¥, Cun A., Yang W, Ellenberg S,, Switzer WM. Chimpanzee adenovirus antibodies in humans, sub- Saharan Africa, Emerging Infect Dis. 2006;12:1596-1599, [PMCID: PMC3290939] [PubMed: 17176582] 75. Fooks A.R. Development of oral vaccines for human use. Curr Opin Mol Ther. 2000;2:00-86. [PubMed: 11249655] 76. Alcock R,, Cottingham M.G, Rollier CS, Furze J, De Costa S.D,, Hanlon M. Long-term thermostabilization of live poxviral and adenoviral vaccine vectors at supraphysiological temperatures in carbohydrate glass. Sei Trans! Med. 2010;2 19ra12, [PubMed: 20371486] 77. Vellinga |, Smith |, Lipiee A, Majhen D., Lemckert A, van Ooij M. Challenges in manufacturing adenoviral vectors for global vaccine product deployment. Hum Gene Ther. 2014;25:318-327, (PubMed: 24593243] 78, Pastoret BP, Brochier B,, Languet B., Thomas 1, Paquot A., Bauduin B. First field trial of fox vaccination against rabies using a vaccinia-rabies recombinant virus. Vet Rec. 1988;123:481-483, [PubMed: 3201693] tps stww.ncb.nim.nih.govlpmetarticles/MC6863069)report=pintable vans. 6122123, 12:05 AM "New human rabies vaccines in the pipeline - PMC 79, Taylor J, Trimarchi C,, Weinberg R., Languet B., Guillemin F, Desmettre P, Efficacy studies on a canarypox-rabies recombinant virus, Vaccine. 1991;9:190-193, [PubMed: 2042391] 80. Cadoz M., Strady A, Meignier B., Taylor ., Tartaglia |, Paoletti rabies glycoprotein, Lancet. 1992;339:1429-1432, [PubMed: 1351126] Immunisation with canarypox virus expressing 81, Weyer J, Rupprecht CE, Mans J, Viljoen GJ., Nel L.lf. Generation and evaluation of a recombinant modified vaccinia virus Ankara vaccine for rabies. Vaccine. 2007;25:4213-4222. [PubMed: 17434244] 82. Gonin P, Oualikene W,, Fournier A, EloitM. Comparison of the efficacy of replication-defective adenovirus and Nyvae poxvirus as vaccine vectors in mice. Vaccine. 1996;14:1083-1087. [PubMed: 8879106] £83, Rupprecht CE, Blass L., Smith K, Orciari L.A., Niezgoda M., Whitfield S.G, Human infection due to recombinant vaccinia-rabies glycoprotein virus. N Engl Med. 2001;345:582-586. [PubMed: 11529212] 84, Chen Z,, Zhou M., Gao X, Zhang G,, Ren G., Gnanadurai CW. A novel rabies vaccine based on a recombinant parainfluenza virus 5 expressing rabies virus glycoprotein. J Virol. 2013;87:2986-2993, [PMCID: PMC3592160] (PubMed: 23269806} 85, Ge|,, Wang X, Tao L,, Wen Z,, Feng N., Yang S, Newcastle disease virus-vectored rabies vaccine is safe, highly immunogenic, and provides long-lasting protection in dogs and cats. J Virol. 2011,88:8241-8252. [PMCID: PMc3147977] [PubMed: 21632762] 86, Yuan Z,, Zhang S, Liu ¥, Zhang F, Fooks A.R,, Li Q. A recombinant pseudorabies virus expressing rabies virus alycoprotein: safety and immunogenicity in dogs. Vaccine, 2008;26:1314~1321. [PubMed: 18262313] 87, Fooks AR, Cliquet &, Finke S, Freuling ©, Hemachudha T,, Mani R.S. Rabies, Nat Rev Dis Primers. 2017;3:17091. (PubMed: 29188797] tps stww.ncb.nim.nih.govlpmetarticles/MC68630691report=pintable 1313

You might also like