<<

Review

RNA-based viral vectors

Expert Rev. Vaccines Early online, 1–30 (2014)

Mark A Mogler1 and The advent of reverse genetic approaches to manipulate the of both positive (+) and Kurt I Kamrud*2 negative (-) sense RNA allowed researchers to harness these genomes for basic research. Manipulation of positive sense RNA genomes occurred first largely because infectious RNA 1Harrisvaccines, Inc., 1102 Southern Hills Drive, Suite 101, Ames, IA 50010, could be transcribed directly from cDNA versions of the RNA genomes. Manipulation of USA negative strand RNA virus genomes rapidly followed as more sophisticated approaches to 2Synthetic Genomics Vaccines, Inc., provide RNA-dependent RNA polymerase complexes coupled with negative-strand RNA 11149 North Torrey Pines, La Jolla, CA templates were developed. These advances have driven an explosion of RNA virus vaccine vector 90237, USA *Author for correspondence: development. That is, development of approaches to exploit the basic replication and expression [email protected] strategies of RNA viruses to produce vaccine antigens that have been engineered into their genomes. This study has led to significant preclinical testing of many RNA virus vectors against a wide range of as well as cancer targets. Multiple RNA virus vectors have advanced through preclinical testing to human clinical evaluation. This review will focus on RNA virus vectors designed to express heterologous genes that are packaged into viral particles and have progressed to clinical testing.

KEYWORDS: • orthomyxovirus • paramyxovirus • reverse genetics • rhabdovirus • RNA virus vector

This review will describe RNA virus vector nsP3 and nsP4) are translated from the genomic development and the reverse genetics approaches RNA. The nsP2 is a viral that processes used to support the development for five RNA the nonstructural polyprotein into its individual virus families: Togaviridae, , Ortho- subunits that function to transcribe negative-

For personal use only. myxoviridae, and Paramyxoviri- sense as well as new positive-sense viral RNA. dae. The RNA vector system(s) designed for The negative-sense replicative intermediate each will be described and a summary of preclin- RNA codes for a subgenomic promoter (26S ical work that has supported advanced testing promoter) that is recognized by the nonstruc- and human clinical evaluation will also be dis- tural and is used to generate a subge- cussed. Although some systems have developed nomic mRNA that represents the 3´ one-third DNA-based vectors, only RNA particle-based of the ; the viral structural proteins are systems will be discussed in this review. Reverse translated from this subgenomic mRNA. Repli- genetics systems for the generation of viruses in cation occurs completely in the of the Arenaviridae family have been described, but cells and progeny viruses bud at the plasma these RNA vectors are still in very early develop- membrane as virus encapsidates Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 ment and therefore will not be examined in this genomic RNA and associates with the envelope review. Finally, reverse genetics approaches to glycoproteins that accumulate on the membrane develop Lentivirus vectors are well established of infected cells [2,3]. but will not be covered here as they have recently Because are positive-sense RNA been reviewed elsewhere [1]. viruses, full-length cDNA clones of their genomes can be used to generate RNA transcripts Togaviridae that, when introduced into susceptible cells, will : Alphavirus initiate a complete virus replication cycle and Alphaviruses have a single-strand, positive-sense generate infectious virus. Manipulation of alpha- RNA genome that carries a 5´ 7-mG cap and a virus genomes is possible through introduction of 3´ polyadenylated region. The alphavirus non- mutations/modifications to the cDNA of their structural proteins are coded for in the 5´ two- respective clones. Two types of alphavirus vectors thirds of the genome and the structural proteins have been developed to express heterologous are coded for in the 3´ one-third of the genome. genes: propagation-competent vectors where a The nonstructural proteins (termed nsP1, nsP2, 26S promoter region is duplicated, either

informahealthcare.com 10.1586/14760584.2015.979798 2014 Informa UK Ltd ISSN 1476-0584 1 Review Mogler & Kamrud

upstream or downstream of a complete structural gene region, that intravenously has been described and preliminary data have drives expression of a foreign gene or propagation-defective repli- been provided that indicate this formulation (1 Â 108 or con vectors where a foreign gene is inserted in place of the struc- 5 Â 109 particles/m2 body surface) was well tolerated and that tural protein gene region [4,5]. The RNA can be packaged a transient increase (five- to tenfold) in IL-12 serum levels was into virus replicon particles (VRP) by supplying the structural pro- detected within the first 3–4 days after injection [16–18]. tein genes in trans. Replicon RNA is packaged into VRP when cells A complete summary of the clinical outcomes of this clinical are cotransfected with helper RNA which encode the full comple- trial has not yet been published. A study protocol for a ment of structural proteins. A split helper system that provides the Phase I/II clinical trial using continuous intratumoral infusion structural proteins on separate helper greatly reduces the of LSFV-IL12 in recurrent glioblastoma multiforme patients chance of an intact genome being regenerated by RNA–RNA has been described, but no data are available on the status of recombination. Thus, the VRP are defective, in that they can infect this clinical trial [18]. target cells in culture or in vivo and can express the foreign gene at high levels, but they lack critical portions of the genome (i.e., the Venezuelan equine vectors structural protein genes) necessary to produce virus particles which Development of VEEV VRP for human clinical evaluation has could spread to other cells [4,5]. Chimeric alphavirus vectors have occurred in both an infectious disease and cancer setting [20–23]. also been developed where the structural region of one alphavirus VEEV VRP vaccines have been tested preclinically against a is exchanged for a different alphavirus structural region; the result- large range of pathogens (TABLE 1), of which only a few have pro- ing chimeric alphavirus is usually attenuated in nature [6–10]. gressed into human clinical evaluation. Some notable VEEV Alphavirus replicon vectors have been developed from a VRP preclinical testing examples include successful vaccines number of viruses within the Togaviridae family; these viruses developed against virus (EBOV), , Lassa consist of Salmonid alphavirus, virus (CHIKV), virus, virus, equine encephalitis viruses, Clostridium Sindbis-like XJ-160, Venezuelan equine encephalitis virus botulinum neurotoxins and anthrax [24–30]; detailed descriptions (VEEV), Sindbis and (SFV), with the of these studies are not provided here in order to focus on majority of vaccine development having been conducted with those VEEV VRP vaccines that have progressed into human VEEV, Sindbis and SFV [4,11–15]. clinical evaluation. Alphavirus vectors have been developed against a large num- The initial human evaluation of a VEEV VRP vaccine was ber of pathogens. A representative list of the many alphavirus with a replicon expressing an HIV antigen carried out by vector particle-based vaccines developed and tested preclinically AlphaVax, Inc. The sequence of an HIV-1 Clade C gag gene is presented in TABLE 1. This review will focus on alphavirus vec- was identified based on a prevalent virus circulating in South For personal use only. tor particle vaccines that have reached human clinical testing or , where one arm of the clinical trial would take have achieved regulatory approval/licensure to highlight how place [31,32]. This first-in-man testing of the VEEV-gag VRP far this vaccine platform has progressed. Clinical evaluation of (AVX101) vaccine demonstrated that the vaccine was safe and VRP vaccines has occurred for a number of infectious diseases well tolerated, but unlike the robust immune responses noted and cancer indications using the VEEV and SFV VRP systems. in preclinical testing of AVX101, anti-Gag immune responses in humans were limited [23]. The AVX101 vaccine expressed a Semliki forest virus vectors nonmyristoylated HIV gag gene and it is known that myristoy- To the best of our knowledge, SFV vectors have only been lation of the Gag protein is important in the production of examined clinically in a cancer setting and with the gene of Gag-derived virus-like particles (VLP) [33,34]. Furthermore, a interest being a cytokine (IL-12). Preclinical studies for the VEEV-gag VRP expressing a myristoylated version of the gag oncolytic nature of SFV supported its use in this setting [16–19]. gene was preliminarily reported to be more immunogenic in Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 A SFV replicon vector expressing murine IL-12 genes was nonhuman than the AVX101 VRP, suggesting that shown to have oncogenic activity when injected directly into the Gag myristoylation mutation (inhibiting Gag-VLP forma- established B16 murine melanoma and P815 mastocytoma tion) may be related to the diminished immune responses tumors to provide support for further development of a SFV noted in the trial [23]. replicon expressing the human cytokine counterpart [16–19]. AlphaVax also developed VEEV VRP vaccines against sea- Ren et al. (2003) encapsulated SFV-IL12 particles in cationic sonal influenza virus and tested them clinically [35,36]. The liposomes (LSFV-IL12) to reduce the possibility of antivector VEEV VRP vaccine for influenza virus was based on a replicon immunity and serum components from negatively affecting vector expressing the A/Wyoming/03/2003 Hemagglutinin antitumor efficacy by inactivating the VRP [18]. The LSFV- (HA) gene that demonstrated promising immunogenicity and IL12 particles injected intraperitoneally in mice implanted sub- protection results from extensive preclinical studies in animal cutaneously with pancreatic cancer cells resulted in reduction of models [37]. The vaccine (AVX502) was tested initially in tumor size if vaccinations were initiated either immediately healthy young adults aged 18–40 years and then in the elderly after tumor implantation or up to 3 weeks after implan- (>65 years); the results of these clinical trials have not yet been tation [18]. A Phase I human clinical trial in kidney car- formally published, but a preliminary summary of the studies cinoma and melanoma patients using LSFV-IL12 delivered is provided [OLMSTEAD R, PERS.COMM.].

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Table 1. Selected RNA virus vector vaccine references. Virus family Subfamily Genus Virus (strain Gene of interest Ref. or modification) Togaviridae Alphavirus SFV HIV gp160 [345] LIV prME and NS1 [346] Influenza A NP and HA [347] HPV16 E6 and E7 [348] proteins HCV core or E2 [349] SIV env, gag-pol, nef, [350] rev and tat HCV nsP3 [351] SINV Plasmodium yoelli CS [352] HTNV, SEOLV S and M [353] HPV 16 E7, fusion E7/ [354] HSP70 RVFV Gn, Gc, nsM [355] HCV C, E1 and E2 [356] VEEV MARV GP, NP, VP40, [25] VP35, VP30 or VP24 BoNT C, MARV GP, [27] Anthrax PA Anthrax PA [28]

For personal use only. Lassa and EBOV GP [29] Influenza A HA [37] VACV, A27L, B5R, [26] A33R, L1R EBOV GP [24] Rhesus [357] lymphocrypticvirus gp350, EBNA-3A, EBNA-3B BVDV E2 [358] Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 EBOV GP [24] Swine influenza A HA, NP [359] VEEV GP, WEEV GP, [30] EEEV GP ANDV: Andes virus; b/hPIV3: Bovine-human parainfluenza virus 3; BoNT-A: Botulinum neurotoxin type A; bPIV3: Bovine parainfluenza virus 3; bRSV: Bovine respira- tory syncytial virus; BVDV: Bovine viral diarrhea virus; CAT: Chloramphenicol acetyltransferase; CEA: Carcinoembryonic antigen; CHIKV: Chikungunya virus; CS: Cir- cumsporozoite; CSFV: Classical swine fever virus; CTL: Cytotoxic T lymphocyte; DEN1, DEN2, DEN3, DEN4: serotypes 1–4; DENV: Dengue virus; EAV: Equine arteritis virus; EBOV: Ebola virus; F: Fusion; G: Attachment glycoprotein; GFP: Green fluorescent protein; GP: Glycoprotein; GPC: Surface glycoprotein precur- sor; HA: Hemagglutinin; hMPV: Human metapneumovirus; HN: Hemagglutinin neuraminidase; hPIV1: Human parainfluenza virus 1; hPIV3: Human parainfluenza virus 3; HPV: Human papilloma virus; hRSV: Human respiratory syncytial virus; HSP70: Heat shock protein 70; HTNV: Hantaan virus; IAV: Influenza A virus; IRES: Internal ribosomal entry site; KUN: ; JEV: virus; LGT: ; LIV: virus; M: Membrane; MARV: Marburg virus; MeV: virus; MS: Middle envelope surface protein; MuV: Mumps virus; MVEV: Murray Valley encephalitis virus; N: Nucleoprotein; NA: Neuraminidase; NDV: Newcastle disease virus; NiV: ; NP: Nucleoprotein; P: Phosphoprotein; prME: Pre-membrane-envelope; RABV: virus; rB/HPIV3: Recombinant bovine- human parainfluenza virus 3; RSV: Respiratory syncytial virus; RVFV: virus; S: Spike; sAg: Surface antigen; SARS-CoV: Severe acute respiratory syn- drome coronavirus; SCFV: Single-cycle flavivirus; SEOLV: Seoul virus; SeV: Sendai virus; SFV: Semliki forest virus; SH: Small hydrophobic protein; SHIV: Simian human immunodeficiency virus; SINV: ; SIV: Simian immunodeficiency virus; SLEV: St Louis encephalitis virus; SV5: Simian virus 5; TBEV: Tick-borne encephalitis virus; VACV: virus; VEEV: Venezuelan equine encephalitis virus; VHSV: Viral hemorrhagic septicemia virus; VSV: Vesicular stomatitis virus;WNV:WestNile virus; YFV: virus.

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

Table 1. Selected RNA virus vector vaccine references (cont.). Virus family Subfamily Genus Virus (strain Gene of interest Ref. or modification) Flaviviridae Flavivirus YFV (17D) JEV prME [45] DEN1–4 prME [49] WNV prME [55] DEN4 (814669 TBEV prME [64] and derivatives) LGT prME [65] SLEV prME [63] WNV prME [70] DEN2 prME [62] DEN2 (PDK-53) DEN1, 3, 4 prME [73] KUN EBOV GP [85] HIV-1 Gag [360] JEV (SA14-14-2) WNV prME [361] DENV prME [87] MVEV (IRES IFN-b [88] attenuated) WNV (SCFV) DENV prME [78] TBEV prME [83] Pestivirus BVDV (CP7) CSFV E2 [91] CSFV E1/E2 [97] For personal use only. BDV E2 [98] BVDV (SD1) GFP [100] BVDV (NADL) Heterologous Erns [101] CSFV BVDV Erns or E2 [105] JEV E(truncated) [104] Influenzavirus Influenza A Circumsporozoite (CS) [119] protein of P. yoelii CS protein of P. [118]

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 falciparum HIV gp41 [122] HIV gp41 [120] HIV GAG [123] ANDV: Andes virus; b/hPIV3: Bovine-human parainfluenza virus 3; BoNT-A: Botulinum neurotoxin type A; bPIV3: Bovine parainfluenza virus 3; bRSV: Bovine respira- tory syncytial virus; BVDV: Bovine viral diarrhea virus; CAT: Chloramphenicol acetyltransferase; CEA: Carcinoembryonic antigen; CHIKV: Chikungunya virus; CS: Cir- cumsporozoite; CSFV: Classical swine fever virus; CTL: Cytotoxic T lymphocyte; DEN1, DEN2, DEN3, DEN4: Dengue virus serotypes 1–4; DENV: Dengue virus; EAV: Equine arteritis virus; EBOV: Ebola virus; F: Fusion; G: Attachment glycoprotein; GFP: Green fluorescent protein; GP: Glycoprotein; GPC: Surface glycoprotein precur- sor; HA: Hemagglutinin; hMPV: Human metapneumovirus; HN: Hemagglutinin neuraminidase; hPIV1: Human parainfluenza virus 1; hPIV3: Human parainfluenza virus 3; HPV: Human papilloma virus; hRSV: Human respiratory syncytial virus; HSP70: Heat shock protein 70; HTNV: Hantaan virus; IAV: Influenza A virus; IRES: Internal ribosomal entry site; KUN: Kunjin virus; JEV: Japanese encephalitis virus; LGT: Langat virus; LIV: Louping ill virus; M: Membrane; MARV: Marburg virus; MeV: Measles virus; MS: Middle envelope surface protein; MuV: Mumps virus; MVEV: Murray Valley encephalitis virus; N: Nucleoprotein; NA: Neuraminidase; NDV: Newcastle disease virus; NiV: Nipah virus; NP: Nucleoprotein; P: Phosphoprotein; prME: Pre-membrane-envelope; RABV: ; rB/HPIV3: Recombinant bovine- human parainfluenza virus 3; RSV: Respiratory syncytial virus; RVFV: Rift Valley fever virus; S: Spike; sAg: Surface antigen; SARS-CoV: Severe acute respiratory syn- drome coronavirus; SCFV: Single-cycle flavivirus; SEOLV: Seoul virus; SeV: Sendai virus; SFV: Semliki forest virus; SH: Small hydrophobic protein; SHIV: Simian human immunodeficiency virus; SINV: Sindbis virus; SIV: Simian immunodeficiency virus; SLEV: St Louis encephalitis virus; SV5: Simian virus 5; TBEV: Tick-borne encephalitis virus; VACV: Vaccinia virus; VEEV: Venezuelan equine encephalitis virus; VHSV: Viral hemorrhagic septicemia virus; VSV: Vesicular stomatitis virus;WNV:WestNile virus; YFV: Yellow fever virus.

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Table 1. Selected RNA virus vector vaccine references (cont.). Virus family Subfamily Genus Virus (strain Gene of interest Ref. or modification) HIV gp160 and Gag [121] hPIV HN [124] RSV F [125] Rhabdoviridae RABV HCV E2 [149] Botulinum neurotoxin [148] SARS-CoV N or S [150] HIV-1 gp160 [153] HIV-1 Gag [154] IL-2 and IL-4 [157] IFN-b [156] SHIV Env and SIV Gag [362] SIV Gag-Pol [158] Vesiculovirus VSV HIV-1 gp120 [164] IAV(H1N1) HA or NA [165] IAV(H5N1) HA [169] HBV MS [171] ANDV GPC [175] HCV C/E1/E2 [176] RSV G or F [174]

For personal use only. Yersinia pestis LcrV [177] Various filovirus GP Reviewed in [180] CD4 [166] CD4 and CXCR4 [167] CHIKV E1/E2 [181] SIV Gag and Env [182] HIV-1 Gag and Env [183] VSV for oncolytic virotherapy Various genes Reviewed in [192]

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 Novirhabdovirus VHSV WNV E [194] Pneumovirinae Pneumovirus hRSV hRSV G (subgroup B) [338] (subgroup A) bRSV hRSV F and/or G [363] ANDV: Andes virus; b/hPIV3: Bovine-human parainfluenza virus 3; BoNT-A: Botulinum neurotoxin type A; bPIV3: Bovine parainfluenza virus 3; bRSV: Bovine respira- tory syncytial virus; BVDV: Bovine viral diarrhea virus; CAT: Chloramphenicol acetyltransferase; CEA: Carcinoembryonic antigen; CHIKV: Chikungunya virus; CS: Cir- cumsporozoite; CSFV: Classical swine fever virus; CTL: Cytotoxic T lymphocyte; DEN1, DEN2, DEN3, DEN4: Dengue virus serotypes 1–4; DENV: Dengue virus; EAV: Equine arteritis virus; EBOV: Ebola virus; F: Fusion; G: Attachment glycoprotein; GFP: Green fluorescent protein; GP: Glycoprotein; GPC: Surface glycoprotein precur- sor; HA: Hemagglutinin; hMPV: Human metapneumovirus; HN: Hemagglutinin neuraminidase; hPIV1: Human parainfluenza virus 1; hPIV3: Human parainfluenza virus 3; HPV: Human papilloma virus; hRSV: Human respiratory syncytial virus; HSP70: Heat shock protein 70; HTNV: Hantaan virus; IAV: Influenza A virus; IRES: Internal ribosomal entry site; KUN: Kunjin virus; JEV: Japanese encephalitis virus; LGT: Langat virus; LIV: Louping ill virus; M: Membrane; MARV: Marburg virus; MeV: Measles virus; MS: Middle envelope surface protein; MuV: Mumps virus; MVEV: Murray Valley encephalitis virus; N: Nucleoprotein; NA: Neuraminidase; NDV: Newcastle disease virus; NiV: Nipah virus; NP: Nucleoprotein; P: Phosphoprotein; prME: Pre-membrane-envelope; RABV: Rabies virus; rB/HPIV3: Recombinant bovine- human parainfluenza virus 3; RSV: Respiratory syncytial virus; RVFV: Rift Valley fever virus; S: Spike; sAg: Surface antigen; SARS-CoV: Severe acute respiratory syn- drome coronavirus; SCFV: Single-cycle flavivirus; SEOLV: Seoul virus; SeV: Sendai virus; SFV: Semliki forest virus; SH: Small hydrophobic protein; SHIV: Simian human immunodeficiency virus; SINV: Sindbis virus; SIV: Simian immunodeficiency virus; SLEV: St Louis encephalitis virus; SV5: Simian virus 5; TBEV: Tick-borne encephalitis virus; VACV: Vaccinia virus; VEEV: Venezuelan equine encephalitis virus; VHSV: Viral hemorrhagic septicemia virus; VSV: Vesicular stomatitis virus;WNV:WestNile virus; YFV: Yellow fever virus.

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

Table 1. Selected RNA virus vector vaccine references (cont.). Virus family Subfamily Genus Virus (strain Gene of interest Ref. or modification) Metapneumovirus hMPV aMPV N or P [319] Paramyxovirinae hPIV3 bPIV3 N [206] hPIV1 HN and F [205] hPIV2 HN and F [205] MeV HA [364] hPIV1 HN and hPIV2 [365] HN and MeV HA Ebo GP [366] bPIV3 N [207] hPIV3/DF-HN Ebo GP [367] hPIV1 hMPV F or G or SH [368] rB/HPIV3 RSV F or RSV G [215] subgroup A RSV F or RSV G [216] subgroup A and B hPIV3 F and HN [207] bPIV3 hPIV3 F and HN [212] (generating b/hPIV3) hPIV3 F and HN [213] (generating rB/HPIV3) b/hPIV3 hPIV3 F and HN [214] For personal use only. RSV F or RSV G or [217] hMPV F hRSV F or soluble [369] hRSV F hMPV F [370] RSV F [371] SeV RSV G [232] RSV F [233] HIV GAG [245] Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 hPIV2 F or hPIV2 HN [228] or HPIV3 HN or RSV F RSV F [229] HIV Env [246] ANDV: Andes virus; b/hPIV3: Bovine-human parainfluenza virus 3; BoNT-A: Botulinum neurotoxin type A; bPIV3: Bovine parainfluenza virus 3; bRSV: Bovine respira- tory syncytial virus; BVDV: Bovine viral diarrhea virus; CAT: Chloramphenicol acetyltransferase; CEA: Carcinoembryonic antigen; CHIKV: Chikungunya virus; CS: Cir- cumsporozoite; CSFV: Classical swine fever virus; CTL: Cytotoxic T lymphocyte; DEN1, DEN2, DEN3, DEN4: Dengue virus serotypes 1–4; DENV: Dengue virus; EAV: Equine arteritis virus; EBOV: Ebola virus; F: Fusion; G: Attachment glycoprotein; GFP: Green fluorescent protein; GP: Glycoprotein; GPC: Surface glycoprotein precur- sor; HA: Hemagglutinin; hMPV: Human metapneumovirus; HN: Hemagglutinin neuraminidase; hPIV1: Human parainfluenza virus 1; hPIV3: Human parainfluenza virus 3; HPV: Human papilloma virus; hRSV: Human respiratory syncytial virus; HSP70: Heat shock protein 70; HTNV: Hantaan virus; IAV: Influenza A virus; IRES: Internal ribosomal entry site; KUN: Kunjin virus; JEV: Japanese encephalitis virus; LGT: Langat virus; LIV: Louping ill virus; M: Membrane; MARV: Marburg virus; MeV: Measles virus; MS: Middle envelope surface protein; MuV: Mumps virus; MVEV: Murray Valley encephalitis virus; N: Nucleoprotein; NA: Neuraminidase; NDV: Newcastle disease virus; NiV: Nipah virus; NP: Nucleoprotein; P: Phosphoprotein; prME: Pre-membrane-envelope; RABV: Rabies virus; rB/HPIV3: Recombinant bovine- human parainfluenza virus 3; RSV: Respiratory syncytial virus; RVFV: Rift Valley fever virus; S: Spike; sAg: Surface antigen; SARS-CoV: Severe acute respiratory syn- drome coronavirus; SCFV: Single-cycle flavivirus; SEOLV: Seoul virus; SeV: Sendai virus; SFV: Semliki forest virus; SH: Small hydrophobic protein; SHIV: Simian human immunodeficiency virus; SINV: Sindbis virus; SIV: Simian immunodeficiency virus; SLEV: St Louis encephalitis virus; SV5: Simian virus 5; TBEV: Tick-borne encephalitis virus; VACV: Vaccinia virus; VEEV: Venezuelan equine encephalitis virus; VHSV: Viral hemorrhagic septicemia virus; VSV: Vesicular stomatitis virus;WNV:WestNile virus; YFV: Yellow fever virus.

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Table 1. Selected RNA virus vector vaccine references (cont.). Virus family Subfamily Genus Virus (strain Gene of interest Ref. or modification) hPIV3 HN [236] Human fibroblast [249] growth factor-2 (FGF- 2) HIV GAG [247] Avulavirus NDV hPIV3 HN [372] hPIV3 or SARS-CoV S [334] Ebo GP [332] Rubulavirus MuV HIV Gag [300] SV5 GFP [299] Morbilivirus MeV MuV HN, F SIV Env, [294] Gag, Pol GFP [253] HIV Env [290] HIV gp140 [289] WNV prM-E or HIV [288] gp140 SIV Gag, Env, Pol/Env, [296] GFP, Lac-Z, CAT WNV prM-E [373] CEA [374] For personal use only. HIV Gag and/or Env [286] CEA [268] H. pylori neutrophil- [375] activating protein HIV p17, p24, RT and [291] Nef fusion HIV p17, p24, RT and [292] Nef fusion RSV F or EBV gp350 [254] Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 HIV Env [295] HIV p17, p24, RT and [293] Nef fusion ANDV: Andes virus; b/hPIV3: Bovine-human parainfluenza virus 3; BoNT-A: Botulinum neurotoxin type A; bPIV3: Bovine parainfluenza virus 3; bRSV: Bovine respira- tory syncytial virus; BVDV: Bovine viral diarrhea virus; CAT: Chloramphenicol acetyltransferase; CEA: Carcinoembryonic antigen; CHIKV: Chikungunya virus; CS: Cir- cumsporozoite; CSFV: Classical swine fever virus; CTL: Cytotoxic T lymphocyte; DEN1, DEN2, DEN3, DEN4: Dengue virus serotypes 1–4; DENV: Dengue virus; EAV: Equine arteritis virus; EBOV: Ebola virus; F: Fusion; G: Attachment glycoprotein; GFP: Green fluorescent protein; GP: Glycoprotein; GPC: Surface glycoprotein precur- sor; HA: Hemagglutinin; hMPV: Human metapneumovirus; HN: Hemagglutinin neuraminidase; hPIV1: Human parainfluenza virus 1; hPIV3: Human parainfluenza virus 3; HPV: Human papilloma virus; hRSV: Human respiratory syncytial virus; HSP70: Heat shock protein 70; HTNV: Hantaan virus; IAV: Influenza A virus; IRES: Internal ribosomal entry site; KUN: Kunjin virus; JEV: Japanese encephalitis virus; LGT: Langat virus; LIV: Louping ill virus; M: Membrane; MARV: Marburg virus; MeV: Measles virus; MS: Middle envelope surface protein; MuV: Mumps virus; MVEV: Murray Valley encephalitis virus; N: Nucleoprotein; NA: Neuraminidase; NDV: Newcastle disease virus; NiV: Nipah virus; NP: Nucleoprotein; P: Phosphoprotein; prME: Pre-membrane-envelope; RABV: Rabies virus; rB/HPIV3: Recombinant bovine- human parainfluenza virus 3; RSV: Respiratory syncytial virus; RVFV: Rift Valley fever virus; S: Spike; sAg: Surface antigen; SARS-CoV: Severe acute respiratory syn- drome coronavirus; SCFV: Single-cycle flavivirus; SEOLV: Seoul virus; SeV: Sendai virus; SFV: Semliki forest virus; SH: Small hydrophobic protein; SHIV: Simian human immunodeficiency virus; SINV: Sindbis virus; SIV: Simian immunodeficiency virus; SLEV: St Louis encephalitis virus; SV5: Simian virus 5; TBEV: Tick-borne encephalitis virus; VACV: Vaccinia virus; VEEV: Venezuelan equine encephalitis virus; VHSV: Viral hemorrhagic septicemia virus; VSV: Vesicular stomatitis virus;WNV:WestNile virus; YFV: Yellow fever virus.

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

The first clinical trial was a placebo-controlled, randomized, and high-dose groups remained neutralizing antibody positive, double-blind study in 216 healthy volunteers (aged 18–40 years), respectively [20]. which evaluated the safety and humoral and cellular immune A Phase I/II study (open-label, dose-escalation study) was responses after one or two inoculations [35]. AVX502 was admin- conducted to evaluate the safety and immunogenicity of carci- istered either subcutaneously or intramuscularly at two dosage noembryonic antigen (CEA(6D))-expressing VRP vaccine levels and was found to be safe and well tolerated irrespective of (AVX701) in patients with CEA-expressing malignancies [21]. the route or the dose given. Both antibody and T- responses The subjects with advanced or metastatic (stage IV) CEA- were efficiently stimulated and persisted for the duration expressing malignancies were treated with one of the three esca- of the 4-month study. Among volunteers with prevaccination lating doses of CEA-expressing VRP. In these subjects, influenza antibody titers (measured by hemagglutination inhibi- AVX701 was administered by intramuscular injection every tion assay) that were below levels thought to be protective, 3 weeks for a minimum of four immunizations, with additional 77 and 80% of these individuals receiving a single low or high doses in patients without progressive disease every 3 months. dose, respectively, responded with protective HAI antibody titers. A total of 28 subjects were treated in the dose-ranging part of A second immunization in these individuals increased seroprotec- the study (dose range: 4 Â 107 IU to 4 Â 108 IU) and no tive responses to 86% for both dosage levels. A rapid and safety or toxicity issues were identified. Following repeated dose-dependent T-cell response (measured by antigen-specific administration, AVX701 was shown to be effective in eliciting IFN-g ELISPOT assay) was also observed and remained signifi- CEA-specific T-cell and antibody response even in the presence cantly elevated for at least 4 months. A second immunization of antivector neutralizing antibodies and elevated Treg levels. extended the duration, but not the magnitude, of these T-cell The study results also suggested that patients with CEA-specific responses. For both antibody and cellular responses, there was T-cell responses exhibited longer overall survival [21]. no significant difference observed between subcutaneous and A VEEV VRP expressing the prostate-specific membrane intramuscular vaccinations. antigen (PSMA) gene was evaluated in a Phase I clinical trial The same VEEV VRP vaccine (AVX502) was also tested in for patients with castration-resistant metastatic prostate can- a Phase I trial involving 28 healthy adults aged 65 years or cer [22]. Two VRP doses (given up to five-times) were tested in older. The trial was a placebo-controlled, randomized, double- patients; the PSMA-VRP was well tolerated at both doses blind study that evaluated responses after administration of tested (9 Â 106 IU or 3.6 Â 107 IU). There did not appear to two doses of vaccine given at a single dosage level. The vac- be clinical benefit at the two PSMA-VRP doses studied sug- cine was safe and well tolerated in this group of healthy, gesting that dosing was suboptimal. This is supported by the ambulatory elderly subjects, paralleling the experience in young positive responses demonstrated with the CEA-VRP vaccine For personal use only. adults. At 4 weeks after the second dose, ten of 20 vaccine described above; the lowest CEA-VRP dose tested was equiva- recipients had a fourfold or greater rise in HAI antibody titer lent to the highest PSMA-VRP dose tested in this study [21,22]. compared to baseline. A significant increase in T-cell responses Two additional cancer trials using VEEV VRP vaccines are was also observed in those receiving vaccine at this time point. currently recruiting patients. A Phase I study using CEA-VRP None of the eight placebo subjects had measurable increases in (AVX701) in patients with stage III colon cancer is now antibody or T-cell responses. The HAI antibody responses recruiting as an extension of the clinical trial already completed measured in the subjects receiving the vaccine exceeded the with this vaccine [40]. Development and testing of a VRP vac- current regulatory approval guidelines for seroconversion in cine for treatment of HER2-neu positive cancers has taken this age group. place [41]. Preclinical testing of neu-VRP in neu transgenic mice A VEEV VRP vaccine expressing human CMV (hCMV) induced robust CD8+, antigen-specific, T cells and tolerance to antigens has been tested in preclinical studies that induced both neu was broken in these animals resulting in the absence or Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 humoral and cellular immune responses to the expressed anti- inhibition of tumor growth in these animals [41]. Based on gens [38,39]. A combination of VRP expressing the extracellular these promising results, a Phase I trial is recruiting patients domain of hCMV glycoprotein B (gB) or a pp65/IE1 fusion with advanced or metastatic HER2-neu (HER2) expressing protein was chosen for human clinical evaluation [39]. Healthy malignancies for treatment with a VRP expressing the extracel- young adults aged 18–45 years were immunized with two dos- lular domain and the transmembrane region of the HER2 gene ages of the vaccine (1 Â 107 or 1 Â 108 infectious units) at 0, (AVX901, [42]). 8 and 24 weeks in a randomized, double-blind Phase I clinical trial [20]. The combination vaccine (AVX601) was well tolerated Licensed veterinary VEEV VRP and all vaccinated individuals developed cellular immune In addition to human clinical testing of VRP vaccines, the responses (by IFN-g ELISPOT) to CMV antigens and poly- VEEV replicon has recently achieved USDA/CVB approval functional CD4+ and CD8+ T cells were detected by polychro- and licensure for veterinary vaccine development. The first matic flow cytometry. After the third dose, 93% of individuals VEEV (strain TC-83) VRP vaccine (expressing the influenza in the low-dose group and all of the individuals in the high- HA gene) against H3N2 swine influenza virus was fully dose group demonstrated CMV neutralizing antibody response; licensed in 2012. Licensure of the H3-VRP vaccine was sup- 6 months after the third dose, 53 and 75% of the low-dose ported by significant safety and efficacy data as well as

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

demonstration of USDA/CVB compliant manufacture of the fever or dengue shock syndrome). Enhancement of dengue vaccine [43]. In addition, conditional licensure of a porcine epi- disease is associated with incomplete protection against one or demic diarrhea virus VRP vaccine as well as an autogenous more serotypes, further complicating dengue vaccine develop- C VRP vaccine have been granted in 2014. ment efforts. The results of the first Phase III clinical trial of the CYD-TDV vaccine were reported in mid-2014 [53] [54]. Flaviviridae The vaccine had statistically significant efficacy against three Genus: Flavivirus serotypes, but not against serotype 2. Despite the relatively Viruses in the genus Flavivirus have nonsegmented, ssRNA(+) poor efficacy against serotype 2, the safety profile of the vac- genomes of approximately 10–12 kb contained within an cine was acceptable and is a promising advance in the field of enveloped, icosahedral nucleocapsid. The genome organization dengue vaccine development. Additional clinical trial results of the is characterized by 5´ and 3´ noncoding are expected in late 2014. regions and a single polyprotein open reading frame. The 5’ The development, safety and preclinical efficacy of a 17D- portion of the polyprotein consists of the three structural pro- based (WNV) vaccine (ChimeriVax-WN02) teins: capsid (C), pre-membrane (prM) and envelope (E), and were recently reviewed [55]. This chimeric virus differs from an the 3´ portion encodes seven nonstructural proteins. The poly- earlier version (ChimeriVax-WN01) by several mutations in protein is co- and post-translationally cleaved into its constitu- the E sequence (L07F, A316V and K440R) that improved ent parts by viral and cellular factors. safety outcomes in neurovirulence studies. The original The Flavivirus genus contains a number of important human has been licensed for use in horses as both a live attenuated and animal pathogens, and vaccine development in this area vaccine and a formalin-inactivated vaccine [55]. has drawn significant attention over the years. Both traditional While most applications of the 17D platform to date have inactivated virus and attenuated virus vaccines have been pro- relied on expression of heterologous prM-E sequence, the duced, with varying degrees of success. Molecular cloning of incorporation of nonflaviviral sequences would potentially allow several flavivirus genomes has enabled the production of recom- for broader applicability of the vaccine platform. Stable incor- binant, chimeric viruses that encode heterologous sequences. In poration of heterologous sequence (e.g., eGFP) between the E most cases, this is accomplished by replacing the backbone and the NS1 sequence has proven to be particularly useful [56]. virus sequence with an orthologous sequence from another fla- Other insertion sites allow for small (~40 codon) insertions vivirus. However, there are some instances where unrelated between the NS2B-NS3 sequences, which enable delivery of T- sequences have been successfully incorporated into functional cell epitopes by this vector [57,58]. flavivirus genomes. The ability to manipulate the genomes of For personal use only. these viruses has proven to be a powerful tool for investigating Dengue virus vectors the biology of both viruses and their hosts. This section will The attenuated Dengue 4 (DEN4) strain 814669 infectious focus on the use of flaviviruses as vaccine platforms for both clone has provided a platform for the creation of numerous related and unrelated viruses. chimeric DEN viruses. The C-prM-E or prM-E sequences derived from Dengue 1 (DEN1), Dengue 2 (DEN2) and Den- Yellow fever virus vectors gue 3 (DEN3) viruses have been used to replace the analogous A system to produce infectious YF-17D virus from in vitro sequence on the DEN4 backbone [59,60]. Further attenuation RNA transcripts has enabled the production of chimeric viruses (in rhesus monkeys and humans) of this and other DENV has that take advantage of the attractive manufacturing and safety been accomplished by deletions in the 3´ UTR as well as other characteristics of the backbone virus [44]. The first chimeric mutations [61,62]. virus based on the 17D backbone replaced the yellow fever The DEN4 backbone containing the prM-E sequence of Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 virus (YFV) prM-E sequence with that of Japanese encephalitis virus (SLEV) demonstrated protective virus (JEV) [45–47]. Clinical development of this vaccine (first immunity in monkeys [63]. This DEN4 platform has also been ChimeriVax-JE; now IMOJEV) has been recently used to express the C-prM-E or prM-E sequence derived from reviewed [48]. tick-borne encephalitis virus (TBEV) and Langat virus Preclinical development of a tetravalent Dengue vaccine (LGT) [64,65]. A Phase I clinical trial of a chimeric LGT/ based on the 17D backbone (ChimeriVax-DEN) demon- DEN4 vaccine resulted in 80% seroconversion to LGT, but strated promising safety and efficacy profiles in nonhuman poor cross-reactivity with wild-type TBEV [66,67]. primates and mice [49]. This vaccine has progressed to human The DEN4 backbone containing the prM-E sequence of clinical trials under the name ‘CYD-TDV’ and is nearing final SLEV demonstrated protective immunity in monkeys [63]. The regulatory approval [50]. A recently completed Phase IIb clini- prM-E of WNV in the DEN4 backbone demonstrated efficacy cal trial showed efficacy against DENV serotypes 1, 3 and against challenge in several preclinical studies [68]. Initial clinical 4 but not against serotype 2 (ClinicalTrials.gov Identi- trials of the ‘WN/DEN4D30’ vaccine candidate induced sero- fier: [51]) [52]. The vaccine itself was well tolerated, and there conversion in 55–75% of vaccinates with a single dose and was no indication that vaccination increased risk for antibody- 89% after two doses [69,70]. The DEN4D30 backbone contains dependent enhancement of infection (dengue hemorrhagic a 30-nucleotide deletion in the 3´ UTR.

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

A DEN2/DEN4 chimera is advancing through clinical trials protective responses in mice, and protected monkeys from chal- as part of the TV003 tetravalent dengue vaccine [62]. Clinical lenge in the case of the chimeric DENV. Another strategy trials have demonstrated good safety and immunogenicity, with involves the attenuation of Murray Valley encephalitis virus 90% of participants seroconverting to at least three dengue (MVEV) by inserting an internal ribosomal entry site between serotypes after a single vaccination [71,72]. the C and PrM sequences [88]. Insertion of sequence encoding Similarly, a backbone based on DEN2 attenuated strain IFN-b downstream of and in frame with the MVEV capsid PDK-53 has been modified to incorporate prM-E sequences sequence was stable using this genome design, and the resulting from DEN1, DEN3 and DEN4 viruses [73]. The four resulting virus exhibited attenuation and immunogenicity in mice. viruses have been evaluated for preclinical safety and efficacy and are advancing as a tetravalent clinical candidate Genus: Pestivirus ‘DENvax’ [74]. A recently completed Phase I clinical trial The pestivirus genome is approximately 12.3 kb of non- showed acceptable safety and immunogenicity, with 62 and segmented ssRNA(+), and the viral particles are enveloped with 96% of participants seroconverting to four or three serotypes, an icosahedral nucleocapsid. The 5´ and 3´ ends of the genome respectively [75,76]. are noncoding, and there is a single open reading frame encod- ing the polyprotein precursor. A 5´ internal ribosomal entry Replication-defective flavivirus vectors site initiates polyprotein , which begins with Npro In contrast to use of replication-competent, chimeric flavivi- (a nonstructural autoprotease), followed by the structural ruses as vaccine vectors, single-cycle flavivirus (SCFV) vectors proteins C, Erns, E1, E2 and finally the remaining nonstruc- have been developed [77–79]. These SCFV are rendered tural proteins p7, NS2–3, NS4A, NS4B, NS5A and NS5B. As replication-defective by an internal deletion in the capsid with the flaviviruses, the polyprotein is co- and post- sequence, which requires trans-complementation with C to translationally cleaved, leading to cytoplasmic replication and recover functional particles. The risk of recombination events eventual virus assembly and release. leading to replication-competent virus was reduced by altera- Viruses of the genus Pestivirus primarily infect ruminants and tions to the cyclization sequence of the trans-complemented C swine, and pestivirus diseases cause significant disruptions to ani- sequence. In the , SCFV expresses subviral particles com- mal agriculture. The type species of the genus, bovine viral diar- posed of the prM-E products, which induce immunity without rhea virus (BVDV), is endemic in many cattle-producing cell-to-cell spread of the vector. The SCFV approach to vaccine regions around the world. Classical swine fever virus (CSFV) is design also allows for substitution of prM-E sequences on a endemic in many countries throughout Asia and , common vector backbone. but has been controlled or eradicated from and For personal use only. Preclinical efficacy of SCFV vaccines has been demonstrated western . Efficacious vaccines against CSFV and BVDV for WNV, DENV, YFV, JEV and TBEV challenge, and a vari- are currently available, but new vaccines that allow differentia- ety of safety studies have been completed [77,78,80–83]. tion of infected and vaccinated animals (DIVA) are desirable as An alternate approach to the development of replication- a means to improve control and eradication efforts [89]. This sec- defective flaviviruses was demonstrated by the packaging of tion will focus primarily on efforts to develop chimeric pestivirus Kunjin virus replicons [84]. In this approach, the sequences vaccines for the prevention of classical swine fever. encoding most of C, and all of prME, are deleted. The repli- con RNA can be modified to accept transgenes in place of the Bovine viral diarrhea virus vectors deleted sequence, and packaging is accomplished by providing Successful recovery of virus from a full-length cDNA clone of the Kunjin virus structural proteins in trans. This system has BVDV isolate CP7 was reported in 1996 [90]. This backbone shown preclinical efficacy in guinea pigs against EBOV chal- was developed as a chimeric vaccine candidate by replacing the Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 lenge and immunogenicity against HIV-1 Gag in mice [85]. E2 sequence of BVDV with the corresponding E2 sequence A method for the production of recombinant YFV using a from CSFV strain Alfort 187 [91]. The resulting chimeric virus, two-component genome strategy has also been developed [86]. CP7_E2alf, has been developed as a potential vaccine allowing This method utilizes intact nonstructural coding sequences for DIVA, with several successful efficacy and safety trials in swine both genomes, but deletes either C or prME. The two genomes vaccination models [92–96]. Another version of this approach is only produce pseuodinfectious particles if both genomes are the CP7_E1E2alf chimeric virus, which has both E1 and present in an infected cell, and can be modified to include E2 sequences derived from CSFV [97]. transgenes (e.g., GFP). Infection of a cell with a single particle Serological cross-reactivity between some pestivirus species will produce subviral particles, transgene products and non- has been exploited to develop novel marker vaccines. The chi- structural proteins but the infection will not propagate. meric pestivirus CP7_E2gif was generated by replacing the BVDV strain CP7 E2 sequence with an analogous E2 sequence Other flavivirus-based vaccine vectors from border disease virus strain Gifhorn [98]. The resultant chi- Recently, the attenuated JEV strain SA14-14-2 was used to meric virus contains no sequence derived from CSFV, but still develop a chimeric virus expressing prME sequences derived induced a protective immune response against CSFV challenge from WNV or DENV [87]. These chimeric vaccines induced and allowed for DIVA serology [98,99].

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

The BVDV genome has also been shown to accept the inser- plasmid DNA engineered to code for a modified influenza A tion of a heterologous gene between the Npro and C sequen- NS segment; the chloramphenicol acetyltransferase (CAT) gene ces. A gene construct encoding green fluorescent protein and was inserted in place of the NS gene and flanked by the wild the 2A protease from foot and mouth disease virus were type 5´ and 3´ noncoding regions. RNA of the engineered inserted in-frame with the polyprotein sequence [100]. Growth CAT segment was produced by in vitro transcription and the kinetics and protein expression were only mildly impacted by RNA was complexed with purified influenza virus polymerase the inserted sequence, suggesting that other heterologous genes complex to form a CAT-RNP. Transfection of the CAT-RNP may be compatible with pestivirus vectors. No in vivo experi- into cells followed by infection with influenza helper virus mentation with this vector design has been reported. resulted in CAT expression in the transfected cells and release Traditional attenuated or inactivated vaccines for BVDV are of influenza virus that had incorporated the CAT RNA seg- not compatible with DIVA serological assays that measure anti- ment. CAT expression was still evident after several passages of body against the Erns antigen. Heterologous pestivirus Erns the modified influenza virus demonstrating that the CAT seg- sequence was used to replace the wild-type Erns sequence in a ment was truly packaged into particles [107]. Enami et al. BVDV backbone [101]. The Erns sequence of giraffe pestivirus, (1990) extended this work by replacing the wild-type influenza reindeer pestivirus and pronghorn antelope pestivirus were A/WSN/33 (WSN) NA gene segment with one containing compatible with the BVDV backbone and produced infectious engineered point mutations by using a WSN helper virus lack- chimeric virus. However, only the pronghorn antelope pestivi- ing a wild-type NA segment, demonstrating the ability to engi- rus/BVDV chimera proved to be distinguishable from BVDV neer the genome of an influenza virus [108]. A rapid expansion infection by anti-Erns serology after vaccination. This approach of methods to produce influenza VLP and to rescue influenza may provide a method to develop DIVA-compatible BVDV from cDNA followed these early successes [109–117]. The culmi- vaccines. nation of the efforts resulted in construction of plasmids con- taining promoter elements to generate both viral RNA (vRNA) Classical swine fever virus vectors transcripts for incorporation into RNP and replication and Deletion of Erns, E2 or partial E2 sequence from a CSFV mRNA transcripts for influenza protein expression to support infectious clone, followed by trans-complementation with the polymerase complex formation and structural protein synthe- missing/defective glycoprotein, allows for recovery of nontrans- sis [111,113,115]. Generation of vRNA with precise 5´ and 3´ ends missible replicon particles [102,103]. These CSFV replicons was accomplished by putting the influenza genome regions induce a protective response in some cases and are compatible under the control of DNA-dependent RNA polymerase I (pol with a DIVA approach to serology. Additionally, E2-deleted I) promoter elements; these RNAs were compatible with RNP For personal use only. CSFV replicons with an inserted sequence derived from JEV formation and replication [115]. Flanking the pol I promoter (truncated E protein) induced immune responses to both elements with DNA-dependent RNA polymerase II promoter CSFV and JEV in both mouse and pig models [104]. elements to produce mRNA for translation supported influenza Chimeric viruses with BVDV Erns or E2 sequence replacing virus protein expression allowing for rescue of recombinant that of CSFV have been shown to protect vaccinated pigs from influenza viruses using only an eight plasmid system [113]. lethal CSFV challenge while maintaining DIVA serological compatibility [93,94,105]. Influenzavirus A vectors With methods in place to manipulate the influenza genome Orthomyxoviridae segments, vaccine antigens were introduced into the vectors for Genus: Influenzavirus a number of targets. A representative list of vaccine targets can Viruses in the Orthomyxoviridae family are negative-sense, sin- be found in TABLE 1. Briefly, influenza vectors have been modi- Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 gle-strand, RNA viruses with eight genome segments (PB2, fied to express circumsporozoite protein B- and T-cell epito- PB1, PA, HA, NP, NA, M and NS). The eight genome seg- pes [118,119], HIV and SIV antigens [120–123], respiratory virus ments express ten viral proteins; M and NS each code for two antigens [124,125] and tumor-associated antigens [126]. proteins generated through differential splicing of the respective In all cases, antigen-specific immune responses were detected in primary transcripts. As is true for viruses in the Paramyxoviri- animal models, demonstrating the potential of influenza virus dae and Rhabdoviridae families, naked genomic RNA is not vectors in a preclinical setting. infectious; only genomic RNA encapsidated with the polymer- Perhaps the most effective use of influenza reverse genetics ase complex (PB1, PB2, PA and NP) to form a ribonucleopro- capabilities has been the development of live attenuated influ- tein (RNP) is able to initiate a replication and transcription enza vaccines used in seasonal influenza vaccination. MedI- cycle. takes place in the nucleus of cells and mmune produced the first cold-adapted live attenuated progeny virus buds from the plasma membrane of infected cells influenza vaccine (termed FluMist in the USA) to be US (reviewed in [106]). FDA licensed for seasonal influenza in 2003 [127]. The reverse The first successful reverse genetics manipulation of any genetically engineered influenza viruses combine attenuating negative-strand RNA virus genome was demonstrated with mutations, cold-adaption and temperature sensitivity character- influenza virus [107]. This was accomplished by generating a istics to restrict vaccine virus replication to the nasopharynx of

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

immunized individuals. Each new seasonal influenza vaccine is increasing the length of the helical nucleoprotein/RNA generated by swapping out relevant HA and NA segments in complex. the context of the attenuated, cold-adapted and temperature- sensitive background influenza segments. FluMist is now Development of reverse genetics systems for approved for seasonal influenza vaccination in persons aged single-strand, negative-sense RNA viruses 2–49 years [128]. Similar to the segmented Orthomyxoviridae viruses, wild-type, The reverse genetics approach to rescue influenza virus pro- naked, negative-sense rhabdovirus RNA is not infectious; rather vides the promise of unprecedented response times to the infectious RNA consists of an RNP complex containing N, P appearance of pandemic influenza virus outbreaks [129]. Advan- and L proteins. Early studies using viral functions expressed ces in synthetic biology have reduced the time required to syn- from plasmid constructs or provided by homologous virus thesize DNA from weeks to hours from receipt of a infected cells resulted in successful encapsidation of small engi- nucleotide sequence [130]. Recently, Dormitzer et al. (2013) neered RNAs or cDNA copies of naturally occurring defective completed a proof-of-concept study to demonstrate the speed interfering RNAs [133–136]. These studies helped define the 3´ with which synthetic seed influenza viruses could be rescued and 5´ minimal terminal sequence requirements for replication completely from cDNA in Madin Darby Canine Kidney of viral RNA. cells [129]. Seed viruses are the source material for amplification Understanding that an RNP complex is required for RNA of traditional inactivated influenza vaccines, so they represent a replication was the key to developing reverse genetics systems for key component in the vaccine production process. rhabdoviruses as well as paramyxoviruses. The basic approach to Dormitzer et al. demonstrated that 4 days and 6.5 h after HA generate recombinant viruses from cDNA for viruses in the and NA sequences were provided to initiate DNA synthesis, Rhabdoviridae and Paramyxoviridae families involved devising synthetic seed viruses could be produced for a putative ways to introduce viral genome (or antigenome) RNA to cells H7N9 pandemic influenza virus [129]. Furthermore, a deriva- that were also expressing the structural proteins N, P and L to tion of the H7N9 synthetic seed virus has been used to manu- reconstitute the active RNP replication complex. This was ini- facture a vaccine suitable for clinical evaluation. A Phase I tially accomplished with the help of a recombinant vaccinia virus clinical trial using the H7N9, MF59 adjuvanted, influenza expressing the bacteriophage T7 DNA-dependent RNA vaccine has been reported (Bart 2014). Eighty-five percent of polymerase [137–142]. Plasmid DNAs coding for a virus genome immunized subjects were immunologically protected (micro- or antigenome and plasmids coding for the N, P and L genes all neutralization titer >1:40) after receiving a second dose of the under the control of a T7 promoter were transfected into cells. MF59 adjuvanted H7N9 inactivated vaccine (Bart 2014) [131]. The transfected cells were then infected with the recombinant For personal use only. The results of this study demonstrate that recombinant influ- T7 expressing vaccinia virus, and a combination of enza virus vaccines represent a powerful tool to rapidly design, T7 transcription terminator sequences and ribozyme elements manufacture and test vaccines in response to a pandemic incorporated into the plasmid DNA constructs resulted in the influenza outbreak. production of authentic or near-authentic viral genome RNAs along with mRNA for each of the critical viral proteins. The Rhabdoviridae T7 transcribed genomic or antigenomic RNA was then recog- Viruses of the family Rhabdoviridae are characteristically rod- nized by the expressed N, P and L proteins to form replication or bullet-shaped enveloped viruses containing a linear, nonseg- complexes and viral replication was initiated resulting in rescue mented, negative-sense ssRNA genome. This section will of viruses completely from cDNA [137–142]. The reverse genetics describe vaccine vector development efforts utilizing rabies virus system was further refined to reduce variables introduced by the (RABV) and vesicular stomatitis virus (VSV). The viral particle vaccinia T7 expressing helper virus as it became known that vac- Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 derives its shape from the helical nucleocapsid protein com- cinia virus enhanced recombination events between genome plexed with genomic RNA. The genome organization of VSV, templates and nonstructural protein helper plasmid con- from 3´ to 5´, consists of nucleocapsid (N), phosphoprotein structs [138]. One approach to reduce this confounding effect was (P), matrix (M), glycoprotein (G) and an RNA-dependent to render the recombinant vaccinia virus incapable of a complete RNA polymerase (L). In the RABV genome, the P protein replication cycle (by UV inactivation) yet allow it to retain ortholog is referred to as NS. Following infection of the host expression of the T7 polymerase [143]. Another approach was to cell, the viral transcriptase complex directs the synthesis of sub- develop continuous cell lines that express the T7 polymerase to genomic mRNAs, pausing at intergenic regions. The transcrip- drive the relevant viral genomes and proteins involved in replica- tase does not proceed in all cases, which manifests as a tion [140,144]. Yet another approach to reduce cell toxicity issues decreasing relative abundance of subgenomic mRNA by gene related to vaccinia helper virus could be attained through addi- position (3´ to 5´) [132]. This characteristic allows for modula- tion of cytosine arabinoside and rifampicin to cell culture during tion of transgene expression by altering the insertion site. The infection [145]. With development of reverse genetics systems genomes of VSV and RABV are amenable to insertion of het- firmly in place, the basics of single-strand, negative-sense RNA erologous genes without drastic impacts on viral replication, virus replication could be explored and these viruses could now and viral particles accommodate the extra genome size by be examined as vaccine vectors.

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Genus: Lyssavirus vectors undergo a single round of replication, but budded prog- Rabies virus vectors eny lack the ability to infect cells [159]. Single-cycle RABV vec- Rabies virus is classified in the genus Lyssavirus and has a tors show great promise as a safe platform for vaccine genome size of approximately 12 kb. The first successful recov- development. Various G-deleted RABV vectors are also being ery of an infectious negative-strand RNA virus entirely from a exploited as tools for the dissection of neuronal function, complete cDNA genome was achieved with RABV [141,146]. although these applications are outside the scope of this Prevalence of antibodies directed against RABV is low among review [152,160]. the human population, except for those who are prophylacti- cally vaccinated due to occupational hazards. Vaccination of Genus: Vesiculovirus companion animals (e.g., dogs) and wildlife for RABV is com- Vesicular stomatitis virus vectors mon, which may limit the utility of these vectors in VSV is an -borne virus that causes periodic epizootics such applications. among domestic livestock. Humans are rarely affected by VSV One strategy for immunization with recombinant RABV and infections are typically asymptomatic. Antibodies to VSV involves the use of inactivated virions that have transgene prod- are uncommon, outside of high-exposure populations (e.g., vet- ucts incorporated into the viral particle. This strategy has been erinarians), making it likely that VSV-based vaccines would used to induce immune responses against hepatitis C virus have little interference from preexisting antivector immunity. E2 glycoprotein and Clostridium botulinum neurotoxin, among A method for producing recombinant VSV (rVSV) was others [147–149]. The E2 glycoprotein of hepatitis C virus was developed shortly after the first infectious clone of RABV was also immunogenic when expressed by replication-competent reported [139,161–163]. Early development efforts demonstrated RABV vectors [149]. The severe acute respiratory syndrome that foreign antigens, such as the envelope protein of HIV-1 or coronavirus (SARS-CoV) nucleocapsid protein or spike glyco- hemagglutinin of influenza virus, could be expressed from protein genes were inserted between the G and L genes of a rVSV vectors [164–167]. Mice were protected from influenza chal- RABV-based vector [150]. Following a single intramuscular lenge following intraperitoneal or intranasal immunization with immunization with the vector expressing spike glycoprotein, rVSV-HA [168]. To reduce vector-associated pathogenesis, two mice developed high levels of SARS-CoV-neutralizing antibod- rVSV vectors were developed with either a C-terminal trun- ies. The use of replication-competent RABV vectors presum- cated G protein or a deleted G protein [161]. The G-deleted ably would be compatible with oral administration, since vector (rVSVDG) protected mice from influenza challenge attenuated RABV vaccines have been used in this manner [151]. without inducing VSV-neutralizing antibodies, but required The development of vaccines targeting HIV-1 has been an two administrations to induce influenza virus-neutralizing anti- For personal use only. active area of RABV vector research [152]. Expression of bodies. In contrast, durable protection of mice against HIV-1 gp160 envelope protein by replication-competent H5N1 influenza challenge was seen even in mice that received RABV induced neutralizing antibodies in mice [153]. Another a single dose of rVSVDG-HA [169]. Further development of the report demonstrated that cytotoxic T lymphocyte (CTL) rVSVDG platform would potentially allow for repeated immu- responses were also primed by this approach [154]. Building on nizations without degrading vaccine vector efficacy [170]. this result, RABV vectors expressing HIV-1 Gag protein were The rVSV platform has demonstrated preclinical efficacy also shown to induce specific CTL responses [155]. Modifica- against a wide range of pathogens, including hepatitis , tions of the RABV G protein, intended to improve safety, did measles virus, sudden acute respiratory syndrome (SARS), respi- not harm the vector’s ability to induce these responses. Rabies ratory syncytial virus (RSV), Andes virus, hepatitis C virus, vectors expressing HIV-1 proteins have also been modified to Nipah virus and Yersinia pestis [171–179]. To our knowledge, simultaneously express cytokines, such as IL-2, IL-4 and none of these vaccine candidates have advanced to human Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 IFN-b [156–158]. Of these trials, IL-2 improved antibody clinical trials at this time. These varied antigens and disease responses slightly, while IFN-b increased CTL responses in models demonstrate the flexibility and potential of VSV as a mice. Simian immunodeficiency virus (SIV) Env and Gag or vaccine platform. Gag-Pol proteins, when expressed by RABV vectors, induced The rVSVDG platform has shown particular utility against neutralizing antibodies and CTL responses in macaques [158]. filoviruses, such as EBOV or Marburg virus (reviewed in [180]). These animals, when challenged with virulent SIV, were pro- Recombinant VSV expressing filovirus glycoprotein have dem- tected from clinical disease. onstrated preventive efficacy in a number of studies in nonhu- Ultimately, the use of RABV-based vectored vaccines in man primates. As a post-exposure therapy, vaccination has humans will depend on the development of constructs with shown mixed results, ranging from 33–100% survival. Certain acceptable safety profiles, particularly neurovirulence. aspects of the laboratory model likely contribute to this vari- Replication-defective and highly attenuated variants are being ability, such as challenge dose or route of exposure. developed, primarily by the deletion of the viral P or M Coincorporation of heterologous proteins into the VSV genes [152]. These viruses have severely reduced fitness and are envelope provide a means to alter the tissue tropism of the primarily of interest as rabies vaccines. However, single-cycle virus, as was demonstrated by incorporation of CD4 and RABV can be produced by deletion of the viral G gene. Such CXCR4 for targeting of HIV-infected cells [166,167]. Expression

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

of CHIKV envelope glycoproteins in place of VSV G, and disease burden falls on young children and the elderly as well without trans-complementation with G, resulted in VSV par- as the immunocompromised. RSV, human metapneumovirus ticles that incorporated CHIKV E1/E2 into the envelope [181]. (hMNV) and human paramyxovirus (hPIV) 1–3 are responsi- This vector induced immune responses specific to CHIKV and ble for a majority of this disease [195–200]. Although live attenu- protected vaccinated mice from challenge. ated vaccines exist for measles virus and mumps virus (also Multiple immunizations of nonhuman primates with rVSV members of the Paramyxoviridae family), RSV, hMNV and expressing Gag and Env protected animals from AIDS follow- hPIV-1–3 have been refractory and no licensed vaccines ing challenge with virulent SIV [182]. Boosting in this study was are available. accomplished by exchanging the serotype of the rVSV G pro- Paramyxoviridae family viruses have a single-strand, negative- tein. Expression of HIV-1 Gag and Env from rVSV stimulated sense RNA genome (ranging from ~13 to 19 kb in size) sur- cytotoxic T-cell responses in mice, consistent with SIV experi- rounded by a lipid envelope; the viruses replicate solely in the ence [183]. Additional trials have supported the use of rVSV vec- cytoplasm of infected cells. All paramyxoviruses code for six tors for the control of HIV in humans [184,185]. Vaccine common genes in their genome; the genes are entitled matrix regimens that utilize rVSV and another vector system in a (M), fusion (F), hemagglutinin neuraminidase (HN; sometimes prime-boost model have also been developed for HIV [186,187]. referred to as G or H), nucleoprotein (NP or N), phosphopro- The first human clinical trial of an rVSV-based vaccine was tein (P) and the large polymerase protein (L). In addition to conducted using a construct encoding HIV-1 Gag [188]. The the six proteins found in all paramyxoviruses, some members dose-escalation study revealed acceptable safety profile, although code for additional proteins. Each gene has precise transcription Gag-specific CD4+ and CD8+ T-cell stimulation was low [189]. initiation and termination signals such that a gradient of The clinical trial is still ongoing at the time of this writing. mRNAs is produced. This occurs because the RNA-dependent The only other report of rVSV use in humans was that of an RNA polymerase pauses as it completes a new mRNA before emergency post-exposure vaccination following a laboratory engaging a new intergenic sequence. If it dissociates from the accident. A laboratory worker was potentially exposed to Zaire genomic RNA before engaging the next start signal, then it ebolavirus (ZEBOV) by accidental needlestick and received must reinitiate at the leader sequence found at the 3´ end of research-grade rVSV expressing the ZEBOV glycoprotein [190]. the genome again. This phenomenon results in those genes at No severe adverse reaction was observed, and the patient the 5´ end of the genome being transcribed into mRNA less showed no signs of ZEBOV infection. Antibodies to VSV and frequently than those at the 3´ end of the genome [133]. ZEBOV were present after vaccination, consistent with the pre- Although not required for efficient replication of all paramyxo- clinical experience with that vector. A Phase I clinical trial of virus RNA templates, the finding that RNA templates with a For personal use only. rVSV expressing ZEBOV glycoprotein is currently recruiting nucleotide length divisible by six (the rule of six) assisted the participants [191]. development of the reverse genetics systems and helped explain The development of rVSV for use in oncolytic virotherapy why natural defective interfering genomes as well as full-length was recently reviewed [192]. The first Phase I clinical trial of viral genomes are generally divisible by a factor of six [200]. rVSV for oncolytic virotherapy has been initiated for treatment The focus of the following section will be on the use of par- of hepatocellular carcinoma [193]. In this case, the virus amyxoviruses as vectors to express heterologous genes that are expresses IFN-b to enhance the safety and efficacy following not naturally associated with the virus that the system is based intratumoral injection. on. This may be a subtle distinction in some of the examples where the heterologous gene is an ortholog of a gene from Genus: Novirhabdovirus another paramyxovirus, but these examples are consistent with Viral hemorrhagic septicemia virus vectors the use of these powerful systems as vectors to express genes Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 Another approach to rhabdovirus-based vaccine development is not naturally associated with the base vector system employed. the use of viruses that do not naturally infect as anti- There are examples of reverse genetically derived paramyxovi- gen presentation vehicles. Viral hemorrhagic septicemia virus, a ruses developed as live attenuated viruses but without engineer- member of the genus Novirhabdovirus, does not replicate at ing expression of heterologous genes; these examples will not temperatures above 20˚C, making it naturally inactivated in be specifically reviewed here. A description of paramyxovirus mammals. Expression of WNV E protein domains by a recom- vector development is subdivided into the genus that the spe- binant viral hemorrhagic septicemia virus resulted in incorpo- cific virus falls into phylogenetically with a general focus on ration of WNV antigen into the viral particles, which were those paramyxovirus vectors that have advanced into human used to immunize mice [194]. Mice challenged with WNV were clinical evaluation. only partially protected, despite the presence of anti-WNV antibodies. Genus: Respirovirus Human parainfluenza virus vectors Paramyxoviridae Recovery of hPIV-1, hPIV-2 and hPIV-3 infectious viruses Viruses in the Paramyxoviridae family are responsible for a sig- completely from cDNA, using either genomic or antigenomic nificant number of respiratory diseases in humans; most of the templates, has been accomplished [201–204]. Soon after the reverse

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

genetics systems were developed, efforts to generate chimeric par- positions affected recombinant virus growth or heterologous ainfluenza viruses began; Tao et al. (1998) replaced the wild-type gene expression [217]. Placement of heterologous genes in the F and HN hPIV-3 genes with those of hPIV-1 to generate a second position was generally superior to the first position [217]. chimeric rPIV3-1 virus [205]. The rPIV3-1 virus demonstrated a The b/hPIV-3 RSV and hMPV recombinant viruses were both mixture of hPIV-3 and hPIV-1 biological characteristics in cell shown to be immunogenic and protective in animal mod- culture. Other chimeric hPIV-3 viruses were generated by els [200]. The b/hPIV-3 RSV F virus (now referred to as replacement of the wild-type hPIV-3 N gene with the N gene MEDI-534) was further developed and tested clinically in from two different strains of bPIV-3 (rHPIV3-NB). The chime- healthy adults, seropositive children aged 1 to 9 years and RSV ric rHPIV3-NB viruses demonstrated restricted replication in and hPIV-3 seronegative children aged 6 to 23 months. Sero- nonhuman primates yet induced robust anti-hPIV-3 immune conversion to RSV and hPIV-3 occurred in 67 and 100% of [206] [200] responses . The rHPIV3-NB chimeric was clinically tested in vaccinated children, respectively . adults and young children as a live attenuated vaccine for hPIV-3; the vaccine was well tolerated and demonstrated Sendai virus vectors restricted replication characteristics similar to those noted in Sendai virus (SeV), also known as murine parainfluenza virus nonhuman primates [207]. type 1, has structural and sequence similarity to hPIV-1 [218,219] and has been shown to be antigenically similar as well [220–222]. Bovine parainfluenza virus vectors For this reason, SeV has been tested as a xenotropic vaccine for Bovine parainfluenza 3 (bPIV-3) has been shown to be antigen- hPIV-1 clinically and has been shown to be well tolerated and ically similar to hPIV-3 and to induce hPIV-3 cross-reactive safe in adults [200]. Recruitment of children and toddlers for and protective immune responses in nonhuman primates [208]. clinical evaluation of SeV (as a vaccine against hPIV-1) in these A bPIV-3 virus (strain Kansas/15626/84) was shown to be well age groups is occurring now [223]. Supported by the clinical tolerated clinically as a live attenuated vaccine for hPIV-3 in safety demonstrated by wild-type SeV it has become the focus children and infants [209–211]. Because of the species-restricted of vaccine vector development. replication noted with bPIV-3 and the clinical safety demon- Garcin et al. (1995) were the first to rescue infectious SeV strated by the live bPIV-3 virus in humans, recombinant from cDNA using vaccinia T7 expressing helper virus and plas- bPIV-3 vectors were developed [212,213]. Haller et al. (2000) mids providing the NP, P and L genes along with the SeV generated a bPIV-3 vector where the F and HN wild-type genome as described above [138]. This was followed closely by genes were replaced with hPIV-3 F and HN genes and the chi- another group in 1996 [145].Inadditiontoreplication- meric was termed b/hPIV3 [212]. A similar bPIV-3-based vector competent SeV vectors, versions modified by deletion of the F For personal use only. was generated by Schmidt et al. (2000) and it was termed gene have been developed that are not transmissible [224].The rBPIV3-FHHNH, where the human PIV-3 F and HN genes SeV-DF vector must be produced in cells that provide F in trans were used to replace the bovine PIV-3 orthologs [213]. This to make particles. These particles are able to infect cells in a [225,226] group also constructed a reciprocal vector, rHPIV3-FBHNB, similar manner to replication-competent SeV and express using hPIV-3 as the base vector modified to contain the bovine heterologous genes but cannot spread to other cells [224,225].Ini- F and HN gene PIV-3 orthologs [213]. tial recombinant SeV viruses expressed reporter genes, but Both the b/hPIV3 and rBPIV-FHHNH (also known as rB/ researchers rapidly began to introduce a host of additional HPIV3) viruses demonstrated replication in vitro that was genes [226,227]. Although SeV has been used to develop vaccines nearly identical to hPIV-3 but reduced replication in vivo in against a large range of pathogens (see TABLE 1), two major areas hamsters and nonhuman primates; both viruses also conferred of SeV vaccine development have emerged; recombinant SeV protection from challenge in vivo similar to that noted in ani- vaccines targeted against respiratory viruses and HIV. These Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 mals after previous infection with hPIV-3 [212–214]. The rB/ areas will be the focus in the following sections. HPIV3 virus was tested in adults, hPIV-3 seropositive children Recombinant SeV vectors expressing the RSV G or F gene and hPIV-3 seronegative children and was shown to be safe have been tested in mice, cotton rats and nonhuman and immunogenic [207]. primates [228–233]. The recombinant SeV RSV vaccines were The rB/HPIV3 vector was engineered to express the G and/ shown to induce both humoral and cellular immune responses or F genes of RSV subgroup A and B as possible vaccines and provided protection from RSV challenge. Importantly, no against both RSV-A and RSV-B and hPIV-3 [215,216]. The six evidence of exacerbation of disease in vaccinated animals was [228–233] new vectors (rB/HPIV3-GA, rB/HPIV3-FA, rB/HPIV3-GAFA, detected after RSV challenge . An important consider- rB/HPIV3-GB, rB/HPIV3-FB and rB/HPIV3-GBFB) were char- ation in RSV vaccine development as vaccine-mediated acterized in vitro and evaluated in hamsters and nonhuman pri- enhancement of disease was noted early in RSV vaccine clinical mates to examine immunogenicity; robust immune responses testing [234,235]. to all three viruses (RSV-A, RSV-B and hPIV-3) were detected. All recombinant SeV are bivalent in nature due to the In parallel, RSV G and F and hMPV F genes were introduced pPIV-1 immunity they provide coupled with expression of a into the b/hPIV-3 vector described above to determine if the heterologous gene. Development of recombinant SeV vaccines location of gene placement in the first or second 3´ genomic that can provide immunity to PIV other than hPIV-1 was a

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

logical extension of this vector. Recombinant SeV expressing Recombinant MeV vectors have been used to express a large the F or HN genes of either hPIV-2 or hPIV-3 have been con- range of proteins (see TABLE 1). Of particular note is their use in structed and tested [228,236,237]. Experiments conducted in cotton both cancer and HIV vaccine development as these vaccines rats demonstrated that the SeV hPIV-2 and hPIV-3 F or HN have advanced into human clinical evaluation. The oncolytic vaccines could induce protective immune responses to their nature of MeV has been linked to changes in receptor tropisms respective hPIV and when combined with recombinant SeV that occur as a result of passage in cell culture to generate RSV-F provided protection from hPIV-1, hPIV-2, hPIV-3 and attenuated vaccine viruses; specifically, attenuated MeV are RSV [228,237]. In addition, recombinant SeV expressing the capable of using CD46 as a cell entry receptor [255,256]. There is influenza HA gene have also been tested and shown to induce an upregulation of CD46 on many tumor cells relative to nor- protection from influenza virus challenge in mice [238]. mal cells so MeV have a natural tropism for cancer cells over Recombinant SeV expressing HIV genes have been tested normal cells [257–264]. Given the long safety history of attenu- extensively and have been shown to induce both cellular and ated MeV vaccines and the fortuitous shift of cell entry recep- humoral immune responses in a range of animal models tor to one predominantly found on many cancer cells, including nonhuman primates [239–246]. A recombinant SeV development of recombinant MeV cancer vaccines was an obvi- expressing the HIV gag gene has been used alone or in combi- ous direction for the system to go. nation with DNA and adenovirus vectors, expressing the same Although attenuated MeV without recombinant genome gene, in a prime-boost vaccination regimen [242,244,245]. Initial manipulation have been clinically tested in cancer patients [265], studies used a replication-competent SeV-gag recombinant vac- the inability to follow the pharmacokinetics of treatment make cine [240,242] and similar studies followed using the SeV DF vec- informed clinical development with oncolytic viruses difficult. tor [244,245]. The replication-competent SeV vaccine, termed In response to this, researchers engineered nonimmunogenic SeV-G (NP), is currently in clinical evaluation in combination soluble marker with no biological function into MeV with an adenovirus 35 vector expressing four HIV genes (gag, vectors. The soluble extracellular domain of human CEA and reverse transcriptase, integrase and nef), termed Ad35-GRIN, the b-subunit of human chorionic gonadotropin (bhCG) were in a prime-boost regimen in Kenya, Rwanda and the chosen as marker peptides and were engineered into recombi- UK [247,248]. Although not discussed in this review, in addition nant MeV [266]. These recombinant MeV can be tracked by fol- to vaccines, recombinant SeV are being used as gene-delivery lowing the kinetics of marker gene expression and this has been vehicles; one example is a nontransmissible SeV expressing the correlated with therapeutic outcomes [266,267]. The MeV-CEA human fibroblast growth factor-2 gene [249]. This recombinant vector retained oncolytic potency in preclinical testing against SeV has been tested clinically in patients with peripheral arte- ovarian and brain cancer and has moved into clinical evaluation For personal use only. rial disease and shown to be well tolerated [249]. Because preex- for each indication ([268–271], ClinicalTrails.gov Identifier: [272]). isting immunity to hPIV-1 can neutralize SeV, the influence of A further refinement of the noninvasive monitoring approach preexisting immunity in human populations to successful SeV was to engineer the human thyroidal sodium iodide symporter vaccine vector development in a clinical setting will ultimately (NIS) membrane ion channel gene into recombinant be determined as more vaccine studies are conducted [250]. MeV [273]. NIS is found on thyroid follicular cells where it functions to transport iodine into cells. Furthermore, NIS Genus: Morbilivirus expression has successfully been exploited in clinical settings for Measles virus vectors thyroid imaging and ablation when combined with radioactive A safe and immunogenic measles virus (MeV) vaccine has iodine [274,275]. By using single photon emission computed been available since 1963 (reviewed in [251]); the original vac- tomography or positron emission tomography imaging and cine strain, Edmonston B, was reactogenic and was eventually radioactive iodine, MeV-NIS infected tissues can be monitored Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 replaced with the Schwarz/Moraten strain, which was derived in a noninvasive manner as infected tissues will concentrate through chicken embryo fibroblast passage of the Edmonston radioactive iodine used as a tracer [276,277]. MeV-NIS has been A and B viruses [252]. In addition to the Schwarz/Moraten used in preclinical noninvasive monitoring against a host of dif- strain, attenuated Edmonston AIK-C and EZ strains are also ferent cancer indications [269,273,276,278,279]. These MeV-NIS pre- used as human vaccines [251]. The MeV vaccine is estimated clinical successes have resulted in a large number of clinical to have been given to more than a billion people and has an trials that are either active, recruiting or about to recruit outstanding history of safety and efficacy [253]. For this reason, patients for the following indications: ovarian cancer, multiple recombinant MeV vaccine vectors enjoy a regulatory advan- myeloma, head and neck cancer and mesothelioma [280–285]. tage over some other paramyxovirus-based vaccine vectors. In addition to using recombinant MeV in cancer virotherapy, The first rescue of MeV completely from cDNA occurred in significant effort has been made in the HIV vaccine field. 1995 and the system was based on the Edmonston B MeV Expression of individual SIV or HIV genes as well as multiple strain [140]. Subsequently, the Schwarz/Moraten and genes individually or as fusion proteins, in a single MeV recom- Edmonston-Zagreb strains of MeV have been rescued from binant, have undergone extensive preclinical testing [286–296]. cDNA and developed as vaccine vectors due to their more One candidate vaccine expresses a fusion protein of p17, p24, attenuated phenotypes [253,254]. reverse transcriptase and Nef (termed MV1-F4) from Clade B

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

HIV-1 [291,292]. The MV1-F4 vaccine has been shown to be of hMPV infections can be dated back to 1958 suggesting that immunogenic, inducing both cellular and humoral responses to many of the cases of bronchiolitis and pneumonia without the F4 antigen, and safe as no toxic effects or virus shedding known etiology may be related to hMPV infection [312,313]. was detected in vaccinated cynomologous macaques [291,292]. Human and avian MPV have been rescued from cDNA and The MV1-F4 vaccine has been tested in a human clinical trial their genomes have also been engineered to express a GFP in young healthy adults aged 18–27 years; the trial is complete reporter gene or for hMPV, extra copies of the hMPV G and but the results have not been made public [297]. F genes [314,315]. Subsequently, recombinant hMPV were gener- ated with either the G or SH or G and SH genes deleted. Genus: Rubulavirus These viruses replicated in cell culture but the DG and DG/D Mumps & parainfluenza virus 5 vectors SH hMPV demonstrated restricted replication in the respira- Two viruses in the Rubulavirus genus have been developed into tory tract of hamsters yet provided protection from wild-type recombinant vaccine vectors, Mumps (MuV) and parainfluenza hMPV replication in their lungs [316]. Additional hMPV dele- virus 5 (PIV5) [298,299]. PIV5 was previously named simian virus tion mutants in the context of DGorDSH backgrounds have 5 (SV5) but in 2009 the International Committee on Taxonomy been generated in the M2 gene, which contains two overlap- of Viruses changed the designation to PIV5 because the virus ping reading frames (M2-1 and M2-2) [317]. Examination of had been isolated from many different species other than mon- replication in the respiratory tract of hamsters and nonhuman keys and the new designation was adopted to remove the primates has demonstrated that hMPV DG and DM2-2 viruses assumption that monkeys were the natural host of the virus. may be promising hMPV vaccines [317,318]. Chimeric hMPV Although the first rescue of MuV occurred in 2000 [298], there viruses incorporating avian MPV N or P orthologs have also [319] has not been significant use of it as a vaccine vector to date. been generated . Both hMPV-NA and hMPV-PA replicated A recombinant MuV vaccine expressing the HIV-1 gag gene was efficiently in cell culture and demonstrated restricted replica- used in a prime-boost vaccination regimen with a recombinant tion in the respiratory tract of nonhuman primates yet pro- [300] [319] VSV expressing the same HIV-1 antigen . The highest Gag- vided protection from hMPV challenge . The hMPV-PA specific T-cell responses were detected when the rMuVgag was chimeric virus is about to undergo clinical testing in adults used as the priming dose followed by the rVSVgag vaccine [300]. aged 18–49 years, hMPV-seropositive children aged 12–59 More preclinical vaccine development has occurred with months, and hMPV-seronegative infants and children aged PIV5 vectors than MuV vectors although no recombinant 6–59 months [320]. PIV5 vectors have progressed past preclinical testing. The first rescue of PIV5 (SV5 at that time) from cDNA occurred in Genus: Avulairus For personal use only. 1997; this group also demonstrated stable expression of a Newcastle disease virus vectors reporter gene (GFP) in recombinant virus passaged up to Newcastle disease virus (NDV) is a worldwide disease of avian 20-times [299] – an important characteristic in replication- species that is capable of causing serious economic losses in the competent vaccine vectors. Mice vaccinated with a recombinant poultry industry [321]. Two groups rescued NDV from cDNA PIV5 vector expressing a model antigen (chicken ovalbumin) separately for the first time in 1999 [322,323]. Prior to reverse were shown to generate high avidity cytotoxic T cells to the genetics approaches to modify NDV, it was known to have antigen equivalent to those induced by a recombinant vaccinia oncolytic potential and has been tested extensively in human virus expressing the same antigen [301]. Arimilli et al. (2008) clinical trials; although oncolytic NDV vectors are being engi- engineered a PIV5 vector to express the TLR5 ligand flagel- neered to express biomolecules to increase their antitumor activ- lin [302]. The recombinant PIV5-flagellin was superior to wild- ity, this review will focus on the infectious disease aspects of type PIV5 at activating IFN-g from both CD8+ and CD4+ NDV vector development. A review of oncolytic NDV vaccine Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 T cells [302]. Subsequently, recombinant PIV5 vectors have been development was recently published and readers are directed to engineered with antigens from vaccinia virus, influenza virus, it for more information on NDV-based cancer treatment [324]. RABV and RSV; all of the recombinant PIV5 vectors demon- NDV strains can have varying virulence and the different strated significant protection from the respective pathogen even virulence categories have been described as velogenic (highly in the face of preexisting PIV5 immunity [303–311]. Interestingly, virulent), mesogenic (intermediate virulence) or lentogenic a recombinant PIV5 vector expressing the H5N1 HA gene (nonvirulent) [321]. Recombinant NDV vaccine vectors have incorporated H5 HA into the recombinant PIV5 virion; inacti- been made from either mesogenic (Beaudette) or lentogenic vated rPIV5-H5 were still capable of protecting mice from (LaSota, Hitchner B1 and Clone-30) NDV strains. In general, challenge, albeit a protective response required a boost of the recombinant NDV vectored vaccines are delivered by the intra- inactivated rPIV5-H5 vaccine [309]. nasal/intratracheal routes of administration; this route induces robust mucosal immune responses that in turn make respira- Genus: Metapneumovirus tory disease targets attractive for NDV vectors. From a veteri- Metapneumovirus vectors nary perspective, all NDV vector vaccines are bivalent in nature Human metapneumovirus is a newly discovered paramyxovi- as immunity to wild-type NDV is provided as a matter of rus [312]. Although only recently described, serological evidence course by the base system in addition to the incorporated

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

heterologous gene [325,326]. Recombinant NDV vaccines against parent virus [338,340–342]. At least two recombinant hRSV vec- influenza virus have been developed with more focus being tors have been developed that express a heterologous placed on viruses with pandemic potential [325–331]. NDV vac- gene [343,344]. The first is a hRSV vector constructed to express cines expressing the HA gene from H5N1 influenza virus have the cystic fibrosis transmembrane conductance regulator and is been tested in mice, chickens and nonhuman primates and being tested as a gene delivery vehicle for treatment of cystic shown to induce robust protective immune responses both in fibrosis [343]. The second is a hRSV replicon that is noncyto- circulation and on mucosal surfaces [326,329,332,333]. Other respi- toxic and capable of long-term gene of interest expression [344]. ratory pathogens have also been targeted for NDV vectored The hRSV replicon is propagation defective, that is, it cannot vaccines [327,334,335]. Recombinant NDV vaccines, based on both make progeny once it enters a cell. The hRSV replicon was Beaudette and LaSota strain backbones, against SARS-CoV and engineered to express both a GFP reporter gene and a blastici- hPIV-3 have been tested in nonhuman primates and shown to din S deaminase (bsd) selectable marker gene [344]. Although be immunogenic and protective [327,334]. A recombinant NDV not tested as a vaccine vector yet, the hRSV replicon has vector expressing the RSV F gene was shown to be more potential for that and potentially as a self-limiting gene deliv- immunogenic than even wild-type RSV infection in a mouse ery vector [344]. model [335]. Nonhuman primates vaccinated by the intranasal/ intratracheal route with a recombinant NDV vector expressing Expert commentary & five-year view the glycoprotein from EBOV demonstrated robust humoral In the nearly 30 years since the development of reverse genetics and cellular immune responses to EBOV glycoprotein, and IgA tools to manipulate RNA virus genomes, this diverse and com- antibody titers were detected in respiratory tract secretions plex group of viruses has become a source of powerful vaccine showing a mucosal immune response [332]. vector systems. This is evidenced by the fact that all five virus HIV has also been a focus of recombinant NDV vaccine devel- families discussed in this review have members that have pro- opment. Early studies incorporated the SIV gag into a recombi- gressed to human clinical evaluation and even to FDA and nant NDV vector [123]. A number or different routes of USDA vaccine licensure. Alphavirus replicon vectors have administration were examined and intranasal NDV-SIVgag shown promise clinically against both infectious disease and administration was found to be superior in a mouse model [123]. cancer targets and fully licensed VEEV VRP vaccines are cur- The optimal site to introduce a HIV-1 gag gene into an NDV rently available for veterinary diseases of swine. Live attenuated vector was determined both in terms of vaccine yield and induc- influenza viruses generated completely from cDNA represent a tion of an immune response [336]. In other NDV-gag vaccine safe and immunogenic alternative to traditionally inactivated studies, the HIV-1 gag gene was modified by fusing it with a vaccines and the same technology, supported by rapid DNA For personal use only. single-chain Fv antibody specific for the dendritic cell receptor, synthesis, has proven ability to produce synthetic seed viruses DEC205. Mice vaccinated with the NDV vaccine expressing the used to produce traditionally inactivated vaccines for pandemic gag-DEC205 Fv fusion induced enhanced CD4+ and CD8+ T- influenza viruses. Vaccine vectors based on recombinant para- cell immune responses [337]. An NDV-gag vaccine was also shown myxoviruses represent real promise for development of poten- to be compatible in a prime-boost vaccine schedule when com- tial RSV vaccines as well as vectors with powerful oncogenic bined with other virus vectors also expressing the gag gene [187]. capacity. Chimeric flaviviruses based on the YFV-17D back- A search of ClinicalTrails.gov indicated that, to date, none of the bone have been approved for use in humans (JEV) and horses infectious disease-specific recombinant NDV vectored vaccines (WNV). The nature of WNV epidemiology presents challenges have entered human clinical evaluation. for clinical efficacy study design and ultimate regulatory approval. Three different tetravalent vaccines for DENV are Genus: Pneumovirus advancing through the clinic, with the CYD-TDV candidate Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 Respiratory syncytial virus vectors now completing Phase III trials. Dengue vaccine candidates Recombinant RSV vectors have been generated from both the based on chimeric DEN/DEN viruses are in Phase I and II tri- human (hRSV) and bovine (bRSV) RSV counter- als. Trials conducted over the next several years will dramati- parts [137,144,338]. Human RSV is a very important pediatric cally advance the practical understanding of DENV respiratory pathogen with impact noted worldwide, yet no vaccinology. Novel replication-defective flaviviruses may pro- licensed vaccine for it is available [339]. This is in large part due vide a tool to improve the safety profile of existing efficacious to clinical testing of an inactivated RSV vaccine in young chil- vaccines. Chimeric pestivirus vaccines show great promise as a dren revealing more severe clinical illness in vaccinated individ- tool for the control of diseases in domestic livestock and wild- uals than unvaccinated individuals who subsequently became life. Vaccines based on recombinant rhabdoviruses have only infected with RSV [234,235]. just begun to enter human clinical trials, initially for HIV. Sev- Development of hRSV or bRSV as a vaccine vector for eral VSV-based preclinical candidates for diseases that represent other pathogens is limited to date. Most recombinant RSV biodefense hazards or public health risks are also being com- viruses tested have been gene deletion or temperature-sensitive mercialized. Oncolytic VSV promises to be an active area of mutant versions of the wild-type virus generated to produce clinical research in the future, with one clinical trial already attenuated live vaccines against either the human or bovine underway.

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Financial & competing interests disclosure includes employment, consultancies, honoraria, stock ownership or options, The authors have no relevant affiliations or financial involvement with expert testimony, grants or patents received or pending, or royalties. any organization or entity with a financial interest in or financial conflict No writing assistance was utilized in the production of this with the subject matter or materials discussed in the manuscript. This manuscript.

Key issues

• Chimeric flaviviruses and live attenuated influenza vaccines (orthomyxoviruses) have achieved regulatory approval for use in humans, while chimeric flaviviruses and alphavirus replicons have reached approval in veterinary animals. • Recombinant alphavirus replicon, chimeric flavivirus, paramyxovirus and vaccines are advancing through human clinical trials supported by preclinical efficacy against a wide variety of infectious disease and cancer targets. • Rational design of flavivirus-, alphavirus- and rhabdovirus-based vaccines has increased vector safety that will support continued testing of these vector platforms. • Most flavivirus and paramyxovirus vector development has focused on the expression of sequences derived from other flaviviruses or paramyxoviruses, respectively; these chimeric vectors offer increased attenuation and great promise for development of vaccines against some of the most important insect-borne and pediatric respiratory illnesses known to man. • The oncolytic nature of many RNA viruses (ex paramyxoviruses and rhabdoviruses) that can be manipulated through reverse genetics represents a significant advancement in cancer treatment. New and continued clinical evaluation of these oncolytic viruses will likely shape the future of cancer therapy.

References aerosol challenge of eastern equine 16. Lundstrom K, Boulikas T. Viral and encephalitis virus. Vaccine 2013;31(11): non-viral vectors in : Papers of special note have been highlighted as: • of interest 1464-70 technology development and clinical trials. •• of considerable interest 9. Wang E, Petrakova O, Adams AP, et al. Technol Cancer Res Treat 2003;2(5): Chimeric Sindbis/eastern equine encephalitis 471-86 1. Emeagi PU, Goyvaerts C, Maenhout S, vaccine candidates are highly attenuated and 17. Lundstrom K. Biology and application of et al. Lentiviral vectors: a versatile tool to immunogenic in mice. Vaccine 2007; alphaviruses in gene therapy. Gene Ther fight cancer. Curr Mol Med 2013;13(4): 25(43):7573-81 2005;12(Suppl 1):S92-97 602-25 For personal use only. 10. Wang E, Volkova E, Adams AP, et al. 18. Ren H, Boulikas T, Lundstrom K, et al. 2. Schlesinger S, Schlesinger MJ. Togaviridae: Chimeric alphavirus vaccine candidates for Immunogene therapy of recurrent the viruses and their replication. In: Fields chikungunya. Vaccine 2008;26(39):5030-9 glioblastoma multiforme with a liposomally Virology. Knipe DM, Howley PM, Editors encapsulated replication-incompetent Lippincott Williams & Wilkins, 11. Glasker S, Lulla A, Lulla V, et al. Virus Semliki forest virus vector carrying the Philadelphia, 2001;p. 895-916 replicon particle based Chikungunya virus neutralization assay using Gaussia luciferase human interleukin-12 gene–a phase I/II 3. Strauss JH, Strauss EG. The alphaviruses: as readout. Virol J 2013;10:235 clinical protocol. J Neurooncol 2003; gene expression, replication, and evolution. 64(1-2):147-54 Microbiol Rev 1994;58(3):491-562 12. Karlsen M, Villoing S, Rimstad E, Nylund A. Characterization of untranslated 19. Zhang J, Asselin-Paturel C, Bex F, et al. 4. Rayner JO, Dryga SA, Kamrud KI. regions of the salmonid alphavirus 3 (SAV3) Cloning of human IL-12 p40 and Alphavirus vectors and vaccination. Rev genome and construction of a SAV3 based p35 DNA into the Semliki Forest virus Med Virol 2002;12(5):279-96 vector: expression of IL-12 in human tumor

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 replicon. Virol J 2009;6:173 5. Vander Veen RL, Harris DL, Kamrud KI. cells. Gene Ther 1997;4(4):367-74 13. Moriette C, Leberre M, Lamoureux A, et al. Alphavirus replicon vaccines. Anim Health Recovery of a recombinant salmonid 20. Bernstein DI, Reap EA, Katen K, et al. Res Rev 2012;13(1):1-9 alphavirus fully attenuated and protective Randomized, double-blind, Phase 1 trial of 6. Atasheva S, Wang E, Adams AP, et al. for rainbow trout. J Virol 2006;80(8): an alphavirus replicon vaccine for Chimeric alphavirus vaccine candidates 4088-98 in CMV seronegative adult protect mice from intranasal challenge with volunteers. Vaccine 2009;28(2):484-93 14. Wolf A, Hodneland K, Frost P, et al. western equine encephalitis virus. Vaccine Salmonid alphavirus-based replicon vaccine 21. Morse MA, Hobeika AC, Osada T, et al. 2009;27(32):4309-19 against infectious salmon anemia (ISA): An alphavirus vector overcomes the presence 7. Paessler S, Fayzulin RZ, Anishchenko M, impact of immunization route and of neutralizing antibodies and elevated et al. Recombinant sindbis/Venezuelan interactions of the replicon vector. Fish numbers of Tregs to induce immune equine encephalitis virus is highly attenuated Shellfish Immunol 2014;36(2):383-92 responses in humans with advanced cancer. and immunogenic. J Virol 2003;77(17): J Clin Invest 2010;120(9):3234-41 15. Wu-yang Z, Guo-dong L. Research on basis 9278-86 •• of reverse genetics system of a Sindbis-like Alphavirus replicon particles break 8. Roy CJ, Adams AP, Wang E, et al. virus XJ-160. Virol J 2011;8:519 tolerance in clinical evaluation and A chimeric Sindbis-based vaccine protects antivector immunity did not impact cynomolgus macaques against a lethal boosting potential.

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

22. Slovin SF, Kehoe M, Durso R, et al. development. AIDS Res Hum 44. Rice CM, Grakoui A, Galler R, A phase I dose escalation trial of vaccine 2003;19(2):133-44 Chambers TJ. Transcription of infectious replicon particles (VRP) expressing 33. Bryant M, Ratner L. Myristoylation- yellow fever RNA from full-length prostate-specific membrane antigen (PSMA) dependent replication and assembly of cDNA templates produced by in vitro in subjects with prostate cancer. Vaccine human immunodeficiency virus 1. Proc Natl ligation. New Biol 1989;1(3):285-96 2013;31(6):943-9 Acad Sci USA 1990;87(2):523-7 45. Chambers TJ, Nestorowicz A, Mason PW, 23. Wecker M, Gilbert P, Russell N, et al. 34. Gottlinger HG, Sodroski JG, Haseltine WA. Rice CM. Yellow fever/Japanese encephalitis Phase I safety and immunogenicity Role of capsid precursor processing and chimeric viruses: construction and biological evaluations of an alphavirus replicon myristoylation in morphogenesis and properties. J Virol 1999;73(4):3095-101 HIV-1 subtype C gag vaccine in healthy infectivity of human immunodeficiency virus 46. Guirakhoo F, Zhang ZX, Chambers TJ, HIV-1-uninfected adults. Clin Vaccine type 1. Proc Natl Acad Sci USA 1989; et al. Immunogenicity, genetic stability, and Immunol 2012;19(10):1651-60 86(15):5781-5 protective efficacy of a recombinant, 24. Herbert AS, Kuehne AI, Barth JF, et al. 35. Phase 1/2 clinical trial of an alphavirus chimeric yellow fever-Japanese encephalitis Venezuelan equine encephalitis virus replicon vaccine for Influenza. Available virus (ChimeriVax-JE) as a live, attenuated replicon particle vaccine protects nonhuman from: http://clinicaltrials.gov/show/ vaccine candidate against Japanese primates from intramuscular and aerosol NCT00440362 encephalitis. Virology 1999;257(2):363-72 challenge with ebolavirus. J Virol 2013; 36. A Safety and immunogenicity trial in adults 47. Monath TP, Soike K, Levenbook I, et al. 87(9):4952-64 65 years of age or over to prevent influenza Recombinant, chimaeric live, attenuated 25. Hevey M, Negley D, Pushko P, et al. (AVX502-003). Available from: http:// vaccine (ChimeriVax) incorporating the Marburg virus vaccines based upon clinicaltrials.gov/show/NCT00706732 envelope genes of Japanese encephalitis alphavirus replicons protect guinea pigs and (SA14-14-2) virus and the capsid and 37. Hubby B, Talarico T, Maughan M, et al. nonhuman primates. Virology 1998;251(1): nonstructural genes of yellow fever (17D) Development and preclinical evaluation of 28-37 virus is safe, immunogenic and protective in an alphavirus replicon vaccine for influenza. non-human primates. Vaccine 1999; 26. Hooper JW, Ferro AM, Golden JW, et al. Vaccine 2007;25(48):8180-9 Molecular smallpox vaccine delivered by 17(15-16):1869-82 38. Reap EA, Dryga SA, Morris J, et al. alphavirus replicons elicits protective 48. Appaiahgari MB, Vrati S. IMOJEV((R)): Cellular and humoral immune responses to immunity in mice and non-human a Yellow fever virus-based novel Japanese alphavirus replicon vaccines expressing primates. Vaccine 2009;28(2):494-511 encephalitis vaccine. Expert Rev Vaccines cytomegalovirus pp65, IE1, and gB 2010;9(12):1371-84 27. Lee JS, Groebner JL, Hadjipanayis AG, proteins. Clin Vaccine Immunol 2007; et al. Multiagent vaccines vectored by 14(6):748-55 49. Guirakhoo F, Pugachev K, Zhang Z, et al. Venezuelan equine encephalitis virus Safety and efficacy of chimeric yellow 39. Reap EA, Morris J, Dryga SA, et al. replicon elicits immune responses to Fever-dengue virus tetravalent vaccine For personal use only. Development and preclinical evaluation of Marburg virus and protection against formulations in nonhuman primates. J Virol an alphavirus replicon particle vaccine for anthrax and botulinum neurotoxin in mice. 2004;78(9):4761-75 cytomegalovirus. Vaccine 2007;25(42): Vaccine 2006;24(47-48):6886-92 7441-9 50. Guy B, Barrere B, Malinowski C, et al. 28. Lee JS, Hadjipanayis AG, Welkos SL. From research to phase III: preclinical, 40. Immunotherapy With CEA(6D) VRP Venezuelan equine encephalitis industrial and clinical development of the Vaccine (AVX701) in patients with stage iii virus-vectored vaccines protect mice against Sanofi Pasteur tetravalent dengue vaccine. colorectal Cancer. Available from: http:// anthrax spore challenge. Infect Immun Vaccine 2011;29(42):7229-41 clinicaltrials.gov/show/NCT01890213 2003;71(3):1491-6 • Reviews the research and clinical 41. Wang X, Wang JP, Maughan MF, 29. Pushko P, Geisbert J, Parker M, et al. development of the chimeric yellow fever Lachman LB. Alphavirus replicon particles Individual and bivalent vaccines based on tetravalent dengue vaccine. containing the gene for HER2/neu inhibit alphavirus replicons protect guinea pigs breast cancer growth and tumorigenesis. 51. Efficacy and safety of dengue vaccine in Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 against infection with Lassa and Ebola Breast cancer research : BCR 2005;7(1): healthy children. Available from: http:// viruses. J Virol 2001;75(23):11677-85 R145-55 clinicaltrials.gov/show/NCT00842530 30. Reed DS, Glass PJ, Bakken RR, et al. 42. A Phase I study to evaluate the antitumor 52. Sabchareon A, Wallace D, Sirivichayakul C, Combined alphavirus replicon particle activity and safety Of AVX901. Available et al. Protective efficacy of the recombinant, vaccine induces durable and cross-protective from: http://clinicaltrials.gov/show/ live-attenuated, CYD tetravalent dengue immune responses against equine NCT01526473 vaccine in Thai schoolchildren: encephalitis viruses. J Virol 2014;88(20): a randomised, controlled phase 2b trial. 12077-86 43. Vander Veen RL, Loynachan AT, Lancet 2012;380(9853):1559-67 Mogler MA, et al. Safety, immunogenicity, 31. Davis NL, West A, Reap E, et al. and efficacy of an alphavirus replicon-based 53. Study of a novel tetravalent dengue vaccine Alphavirus replicon particles as candidate swine influenza virus hemagglutinin vaccine. in healthy children aged 2 zto 14 years in HIV vaccines. IUBMB Life 2002;53(4-5): Vaccine 2012;30(11):1944-50 asia. Available from: http://clinicaltrials.gov/ 209-11 show/NCT01373281 • The first alphavirus replicon particle 32. Williamson C, Morris L, Maughan MF, 54. Capeding MR, Tran NH, Hadinegoro SR, et al. Characterization and selection of vaccine fully licensed by the USDA for et al. Clinical efficacy and safety of a novel HIV-1 subtype C isolates for use in vaccine veterinary use. tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised,

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

observer-masked, placebo-controlled trial. encephalitis virus. Virology 2000;274(1): phase 1 study. The Lancet Infect Dis 2014; Lancet 2014;384(9951):1358-65 26-31 14(9):830-8 55. Dayan GH, Pugachev K, Bevilacqua J, et al. 66. Safety of and immune response to a 77. Mason PW, Shustov AV, Frolov I. Preclinical and clinical development of a tick-borne encephalitis vaccine (LGT Production and characterization of vaccines YFV 17 D-based chimeric vaccine against (TP21)/DEN4) in Healthy Adults. Available based on flaviviruses defective in replication. West Nile virus. Viruses 2013;5(12): from: http://clinicaltrials.gov/show/ Virology 2006;351(2):432-43 3048-70 NCT00118924 78. Suzuki R, Winkelmann ER, Mason PW. 56. Martins MA, Bonaldo MC, Rudersdorf RA, 67. Wright PF, Ankrah S, Henderson SE, et al. Construction and characterization of a et al. Immunogenicity of seven new Evaluation of the Langat/dengue 4 chimeric single-cycle chimeric flavivirus vaccine recombinant yellow fever viruses 17D virus as a live attenuated tick-borne candidate that protects mice against lethal expressing fragments of SIVmac239 Gag, encephalitis vaccine for safety and challenge with dengue virus type 2. J Virol Nef, and Vif in Indian rhesus macaques. immunogenicity in healthy adult volunteers. 2009;83(4):1870-80 PLoS One 2013;8(1):e54434 Vaccine 2008;26(7):882-90 79. Widman DG, Frolov I, Mason PW. 57. Barba-Spaeth G, Longman RS, Albert ML, 68. Pletnev AG, Swayne DE, Speicher J, et al. Third-generation flavivirus vaccines based Rice CM. Live attenuated yellow fever 17D Chimeric West Nile/dengue virus vaccine on single-cycle, encapsidation-defective infects human DCs and allows for candidate: preclinical evaluation in mice, viruses. Adv Virus Res 2008;72:77-126 presentation of endogenous and geese and monkeys for safety and 80. Ishikawa T, Widman DG, Bourne N, et al. recombinant T cell epitopes. J Exp Med immunogenicity. Vaccine 2006;24(40-41): Construction and evaluation of a chimeric 2005;202(9):1179-84 6392-404 pseudoinfectious virus vaccine to prevent 58. Tao D, Barba-Spaeth G, Rai U, et al. 69. Safety of and immune response to a west Japanese encephalitis. Vaccine 2008;26(22): Yellow fever 17D as a vaccine vector for nile virus vaccine (WN/DEN4delta30) in 2772-81 microbial CTL epitopes: protection in a Healthy Adults. Available from: http:// 81. Nelson MH, Winkelmann E, Ma Y, et al. malaria model. J Exp Med 2005; clinicaltrials.gov/show/NCT00537147 Immunogenicity of RepliVAX WN, a novel 201(2):201-9 70. Durbin AP, Wright PF, Cox A, et al. The single-cycle West Nile virus vaccine. Vaccine 59. Bray M, Lai CJ. Construction of intertypic live attenuated chimeric vaccine rWN/ 2010;29(2):174-82 chimeric dengue viruses by substitution of DEN4Delta30 is well-tolerated and 82. Widman DG, Ishikawa T, Fayzulin R, et al. structural protein genes. Proc Natl Acad Sci immunogenic in healthy flavivirus-naive Construction and characterization of a USA 1991. 88(22):10342-6 adult volunteers. Vaccine 2013;31(48): second-generation pseudoinfectious West 60. Chen W, Kawano H, Men R, et al. 5772-7 Nile virus vaccine propagated using a new Construction of intertypic chimeric dengue 71. Evaluation of the safety and immune cultivation system. Vaccine 2008;26(22): viruses exhibiting type 3 antigenicity and response of five admixtures of a Tetravalent 2762-71

For personal use only. neurovirulence for mice. J Virol 1995;69(8): Dengue Virus Vaccine. Available from: 83. Rumyantsev AA, Goncalvez AP, 5186-90 http://clinicaltrials.gov/show/NCT01072786 Giel-Moloney M, et al. Single-dose vaccine 61. Blaney JE Jr., Durbin AP, Murphy BR, 72. Durbin AP, Kirkpatrick BD, Pierce KK, against tick-borne encephalitis. Proc Natl Whitehead SS. Development of a live et al. A single dose of any of four different Acad Sci USA 2013;110(32):13103-8 attenuated dengue virus vaccine using live attenuated tetravalent dengue vaccines is 84. Khromykh AA, Varnavski AN, reverse genetics. Viral Immunol 2006;19(1): safe and immunogenic in flavivirus-naive Westaway EG. Encapsidation of the 10-32 adults: a randomized, double-blind clinical flavivirus kunjin replicon RNA by using a 62. Durbin AP, Kirkpatrick BD, Pierce KK, trial. J Infect Dis 2013;207(6):957-65 complementation system providing Kunjin et al. Development and clinical evaluation 73. Huang CY, Butrapet S, Tsuchiya KR, et al. virus structural proteins in trans. J Virol of multiple investigational monovalent Dengue 2 PDK-53 virus as a chimeric 1998;72(7):5967-77 DENV vaccines to identify components for carrier for tetravalent dengue vaccine 85. Reynard O, Mokhonov V, Mokhonova E, inclusion in a live attenuated tetravalent development. J Virol 2003;77(21):11436-47 et al. Kunjin virus replicon-based vaccines Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 DENV vaccine. Vaccine 2011;29(42): 74. Osorio JE, Huang CY, Kinney RM, expressing Ebola virus glycoprotein GP 7242-50 Stinchcomb DT. Development of DENVax: protect the guinea pig against lethal Ebola 63. Blaney JE Jr, Speicher J, Hanson CT, et al. a chimeric dengue-2 PDK-53-based virus infection. J Infect Dis 2011; Evaluation of St. Louis encephalitis virus/ tetravalent vaccine for protection against 204(Suppl 3):S1060-5 dengue virus type 4 antigenic chimeric . Vaccine 2011;29(42):7251-60 86. Shustov AV, Mason PW, Frolov I. viruses in mice and rhesus monkeys. 75. Safety and immunogenicity study to assess Production of pseudoinfectious 2008;26(33):4150-9 denvax, a live attenuated Tetravalent virus with a two-component genome. J 64. Pletnev AG, Bray M, Huggins J, Lai CJ. Vaccine for Prevention of Dengue Fever. Virol 2007;81(21):11737-48 Construction and characterization of Available from: http://clinicaltrials.gov/show/ 87. Li XF, Deng YQ, Yang HQ, et al. chimeric tick-borne encephalitis/dengue NCT01224639 A chimeric dengue virus vaccine using type 4 viruses. Proc Natl Acad Sci USA 76. Osorio JE, Velez ID, Thomson C, et al. Japanese encephalitis virus vaccine strain 1992;89(21):10532-6 Safety and immunogenicity of a SA14-14-2 as backbone is immunogenic 65. Pletnev AG, Karganova GG, Dzhivanyan TI, recombinant live attenuated tetravalent and protective against either parental virus et al. Chimeric Langat/Dengue viruses dengue vaccine (DENVax) in in mice and nonhuman primates. J Virol protect mice from heterologous challenge flavivirus-naive healthy adults in Colombia: 2013;87(24):13694-705 with the highly virulent strains of tick-borne a randomised, placebo-controlled,

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

88. Frese M, Lee E, Larena M, et al. Internal chimeric pestivirus strain CP7_E2gif. Vet 110. Enami M, Palese P. High-efficiency entry site-based attenuation of a Microbiol 2014;170(3-4):224-31 formation of influenza virus transfectants. flavivirus candidate vaccine and evaluation 100. Fan ZC, Dennis JC, Bird RC. Bovine viral J Virol 1991;65(5):2711-13 of the effect of beta coexpression diarrhea virus is a suitable viral vector for 111. Fodor E, Devenish L, Engelhardt OG, et al. on vaccine properties. J Virol 2014;88(4): stable expression of heterologous gene when Rescue of influenza A virus from 2056-70 inserted in between N(pro) and C genes. recombinant DNA. J virol 1999;73(11): 89. Newcomer BW, Givens MD. Approved and Virus Res 2008;138(1-2):97-104 9679-82 experimental countermeasures against 101. Luo Y, Yuan Y, Ankenbauer RG, et al. 112. Gomez-Puertas P, Mena I, Castillo M, et al. pestiviral diseases: bovine viral diarrhea, Construction of chimeric bovine viral Efficient formation of influenza virus-like classical swine fever and border disease. diarrhea viruses containing glycoprotein E particles: dependence on the expression Antiviral Res 2013;100(1):133-50 rns of heterologous pestiviruses and levels of viral proteins. J Gen Virol 1999; 90. Meyers G, Tautz N, Becher P, et al. evaluation of the chimeras as potential 80(Pt 7):1635-45 Recovery of cytopathogenic and marker vaccines against BVDV. Vaccine 113. Hoffmann E, Neumann G, Kawaoka Y, noncytopathogenic bovine viral diarrhea 2012;30(26):3843-8 et al. A DNA transfection system for viruses from cDNA constructs. J Virol 102. Widjojoatmodjo MN, van Gennip HG, generation of influenza A virus from eight 1996;70(12):8606-13 Bouma A, et al. Classical swine fever virus E plasmids. Proc Natl Acad Sci USA 2000; 91. Reimann I, Depner K, Trapp S, Beer M. (rns) deletion mutants: 97(11):6108-13 An avirulent chimeric Pestivirus with altered trans-complementation and potential use as 114. Mena I, Vivo A, Perez E, Portela A. Rescue cell tropism protects pigs against lethal nontransmissible, modified, live-attenuated of a synthetic chloramphenicol infection with classical swine fever virus. marker vaccines. J Virol 2000;74(7): acetyltransferase RNA into influenza Virology 2004;322(1):143-57 2973-80 virus-like particles obtained from 92. Koenig P, Lange E, Reimann I, 103. van Gennip HG, Bouma A, van Rijn PA, recombinant plasmids. J Virol 1996;70(8): Beer M. CP7_E2alf: a safe and efficient et al. Experimental non-transmissible marker 5016-24 marker vaccine strain for oral immunisation vaccines for classical swine fever (CSF) by 115. Neumann G, Watanabe T, Ito H, et al. of wild boar against Classical swine fever trans-complementation of E(rns) or E2 of Generation of influenza A viruses entirely virus (CSFV). Vaccine 2007;25(17):3391-9 CSFV. Vaccine 2002;20(11-12):1544-56 from cloned cDNAs. Proc Natl Acad Sci 93. Blome S, Aebischer A, Lange E, et al. 104. Yang Z, Wu R, Li RW, et al. Chimeric USA 1999;96(16):9345-50 Comparative evaluation of live marker classical swine fever (CSF)-Japanese 116. Neumann G, Watanabe T, Kawaoka Y. vaccine candidates "CP7_E2alf" and "flc11" encephalitis (JE) viral replicon as a Plasmid-driven formation of influenza along with C-strain "Riems" after oral non-transmissible vaccine candidate against virus-like particles. J Virol 2000;74(1): vaccination. Veterinary microbiology 2012; CSF and JE infections. Virus Res 2012; 547-51 158(1-2):L:42-59 165(1):61-70 For personal use only. 117. Neumann G, Zobel A, Hobom G. RN 94. Eble PL, Geurts Y, Quak S, et al. Efficacy 105. van Gennip HG, van Rijn PA, A polymerase I-mediated expression of of chimeric Pestivirus vaccine candidates Widjojoatmodjo MN, et al. Chimeric influenza viral RNA molecules. Virology against classical swine fever: protection and classical swine fever viruses containing 1994;202(1):477-9 DIVA characteristics. Veterinary envelope protein E(RNS) or E2 of bovine 118. Miyahira Y, Garcia-Sastre A, Rodriguez D, microbiology 2013;162(2-4):437-46 viral diarrhoea virus protect pigs against et al. Recombinant viruses expressing a challenge with CSFV and induce a 95. Renson P, Le Dimna M, Keranflech A, human malaria antigen can elicit potentially distinguishable antibody response. Vaccine et al. CP7_E2alf oral vaccination confers protective immune CD8+ responses in 2000;19(4-5):447-59 partial protection against early classical mice. Proc Natl Acad Sci USA 1998;95(7): swine fever virus challenge and interferes 106. Krug R, Alonso-Kaplen FV, Julkunen I, 3954-9 with pathogeny-related cytokine responses. Katze MG. Expression and replication of 119. Rodrigues M, Li S, Murata K, et al. Vet Res 2013;44:9 the influenza virus genome. In: The Influenza and vaccinia viruses expressing Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 influenza viruses. Krug R, Editor Plenum 96. Feliziani F, Blome S, Petrini S, et al. First malaria CD8+ T and B cell epitopes. Press; New York, NY: 1989. p. 89-152 assessment of classical swine fever marker Comparison of their immunogenicity and vaccine candidate CP7_E2alf for oral 107. Luytjes W, Krystal M, Enami M, et al. capacity to induce protective immunity. immunization of wild boar under field Amplification, expression, and packaging of J Immunol 1994;153(10):4636-48 conditions. Vaccine 2014;32(18):2050-5 foreign gene by influenza virus. Cell 1989; 120. Ferko B, Katinger D, Grassauer A, et al. 59(6):1107-13 97. Reimann I, Depner K, Utke K, et al. Chimeric influenza virus replicating Characterization of a new chimeric marker 108. Enami M, Luytjes W, Krystal M, Palese P. predominantly in the murine upper vaccine candidate with a mutated antigenic Introduction of site-specific mutations into respiratory tract induces local immune E2-epitope. Vet Microbiol 2010;142(1-2): the genome of influenza virus. Proc Natl responses against human immunodeficiency 45-50 Acad Sci USA 1990;87(10):3802-5 virus type 1 in the genital tract. J Infect Dis 98. Rasmussen TB, Uttenthal A, Reimann I, 109. Enami M, Enami K. Characterization of 1998;178(5):1359-68 et al. Virulence, immunogenicity and influenza virus NS1 protein by using a 121. Garulli B, Di Mario G, Stillitano MG, vaccine properties of a novel chimeric novel helper-virus-free reverse genetic et al. Exploring mucosal immunization with pestivirus. J Gen Virol 2007;88(Pt 2):481-6 system. J Virol 2000;74(12):5556-61 a recombinant influenza virus carrying an 99. von Rosen T, Rangelova D, Nielsen J, et al. HIV-polyepitope in mice with pre-existing DIVA vaccine properties of the live

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

immunity to influenza. Vaccine 2014; Lippincott-Raven Publishers; Philidelphia, 144. Buchholz UJ, Finke S, Conzelmann KK. 32(21):2501-6 PA: 1996. P. 561-75 Generation of bovine respiratory syncytial 122. Muster T, Ferko B, Klima A, et al. Mucosal 133. Lamb RAaK D. Paramyxoviridae: The virus (BRSV) from cDNA: BRSV NS2 is model of immunization against human viruses and their replication. In: not essential for virus replication in tissue immunodeficiency virus type 1 with a Fundamental Virology. Fields BN, culture, and the human RSV leader region chimeric influenza virus. J Virol 1995; Knipe PM and Howly PM, Editors acts as a functional BRSV genome 69(11):6678-86 Lippincott-Raven Publishers; Philadelphia, promoter. J Virol 1999;73(1):251-9 123. Nakaya Y, Zheng H, Garcia-Sastre A. PA: 1996. p. 577-604 145. Kato A, Sakai Y, Shioda T, et al. Initiation Enhanced cellular immune responses to SIV 134. Pattnaik AK, Ball LA, LeGrone AW, of Sendai virus multiplication from Gag by immunization with influenza and Wertz GW. Infectious defective interfering transfected cDNA or RNA with negative or vaccinia virus recombinants. Vaccine 2003; particles of VSV from transcripts of a positive sense. Genes Cells 1996;1(6): 21(17-18):2097-106 cDNA clone. Cell 1992;69(6):1011-20 569-79 124. Maeda Y, Hatta M, Takada A, et al. Live 135. Pattnaik AK, Wertz GW. Replication and 146. Conzelmann KK, Schnell M. Rescue of bivalent vaccine for parainfluenza and amplification of defective interfering particle synthetic genomic RNA analogs of rabies influenza virus infections. J Virol 2005; RNAs of vesicular stomatitis virus in cells virus by plasmid-encoded proteins. J Virol 79(11):6674-9 expressing viral proteins from vectors 1994;68(2):713-19 125. Zhang P, Gu H, Bian C, et al. containing cloned cDNAs. J Virol 1990; 147. Koser ML, McGettigan JP, Tan GS, et al. Characterization of recombinant influenza 64(6):2948-57 Rabies virus nucleoprotein as a carrier for A virus as a vector expressing respiratory 136. Pattnaik AK, Wertz GW. Cells that express foreign antigens. Proc Natl Acad Sci USA syncytial virus fusion protein epitopes. all five proteins of vesicular stomatitis virus 2004;101(25):9405-10 J Gen Virol 2014;95(Pt 9):1886-91 from cloned cDNAs support replication, 148. Mustafa W, Al-Saleem FH, Nasser Z, et al. 126. Strobel I, Krumbholz M, Menke A, et al. assembly, and budding of defective Immunization of mice with the non-toxic Efficient expression of the tumor-associated interfering particles. Proc Natl Acad Sci HC50 domain of botulinum neurotoxin antigen MAGE-3 in human dendritic cells, USA 1991;88(4):1379-83 presented by rabies virus particles induces a using an avian influenza virus vector. Hum 137. Collins PL, Hill MG, Camargo E, et al. strong immune response affording Gene Ther 2000;11(16):2207-18 Production of infectious human respiratory protection against high-dose botulinum neurotoxin challenge. Vaccine 2011;29(28): 127. Harper SA, Fukuda K, Cox NJ, Bridges CB syncytial virus from cloned cDNA confirms 4638-45 . Advisory Committee on Immunization P. an essential role for the transcription ’ Using live, attenuated influenza vaccine for elongation factor from the 5 proximal open 149. Siler CA, McGettigan JP, Dietzschold B, prevention and control of influenza: reading frame of the M2 mRNA in gene et al. Live and killed rhabdovirus-based supplemental recommendations of the expression and provides a capability for vectors as potential hepatitis C vaccines. vaccine development. Proc Natl Acad Sci Virology 2002;292(1):24-34 For personal use only. Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep USA 1995;92(25):11563-7 150. Faber M, Lamirande EW, Roberts A, et al. 2003;52(RR-13):1-8 138. Garcin D, Pelet T, Calain P, et al. A highly A single immunization with a 128. Mallory RM, Yi T, Ambrose CS. Shedding recombinogenic system for the recovery of rhabdovirus-based vector expressing severe of Ann Arbor strain live attenuated infectious Sendai paramyxovirus from acute respiratory syndrome coronavirus influenza vaccine virus in children cDNA: generation of a novel copy-back (SARS-CoV) S protein results in the 6-59 months of age. Vaccine 2011;29(26): nondefective interfering virus. EMBO J production of high levels of 4322-7 1995;14(24):6087-94 SARS-CoV-neutralizing antibodies. J Gen Virol 2005;86(Pt 5):1435-40 129. Dormitzer PR, Suphaphiphat P, 139. Lawson ND, Stillman EA, Whitt MA, Gibson DG, et al. Synthetic generation of Rose JK. Recombinant vesicular stomatitis 151. Rupprecht CE, Hanlon CA, Blanton J, influenza vaccine viruses for rapid response viruses from DNA. Proc Natl Acad Sci et al. Oral vaccination of dogs with to pandemics. Sci Transl Med 2013;5(185): USA 1995;92(10):4477-81 recombinant rabies virus vaccines. Virus Res Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 185ra168 140. Radecke F, Spielhofer P, Schneider H, et al. 2005;111(1):101-5 •• Synthetic generation of influenza vaccine Rescue of measles viruses from cloned 152. Gomme EA, Wanjalla CN, Wirblich C, viruses for rapid response to pandemics. DNA. The EMBO journal 1995;14(23): Schnell MJ. Rabies virus as a research tool 5773-84 and viral vaccine vector. Adv Virus Res 130. Gibson DG. Enzymatic assembly of 2011;79:139-64 overlapping DNA fragments. Methods 141. Schnell MJ, Mebatsion T, Conzelmann KK. Enzymol 2011;498:349-61 Infectious rabies viruses from cloned cDNA. 153. Schnell MJ, Foley HD, Siler CA, et al. EMBO J 1994;13(18):4195-203 Recombinant rabies virus as potential 131. Bart SA, Hohenboken M, Della Cioppa G, 142. Whelan SP, Ball LA, Barr JN, Wertz GT. live-viral vaccines for HIV-1. Proc Natl et al. A cell culture-derived MF59- Acad Sci USA 2000;97(7):3544-9 adjuvanted pandemic A/H7N9 vaccine is Efficient recovery of infectious vesicular immunogenic in adults. Sci Transl Med stomatitis virus entirely from cDNA clones. 154. McGettigan JP, Sarma S, Orenstein JM, 2014;6(234):234ra255 Proc Natl Acad Sci USA 1995;92(18): et al. Expression and immunogenicity of 8388-92 human immunodeficiency virus type 1 Gag 132. Wagner RRaR JK. Rhabdoviridae: The expressed by a replication-competent viruses and their replication. In: 143. Tsung K, Yim JH, Marti W, et al. Gene expression and of vaccinia rhabdovirus-based vaccine vector. J Virol Fundamental Virology. Fields BN, 2001;75(18):8724-32 Knipe PM. and Howly PM, Editors virus inactivated by psoralen and long-wave UV light. J Virol 1996;70(1):165-71

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

155. McGettigan JP, Foley HD, Belyakov IM, Proc Natl Acad Sci USA 1996;93(21): challenge for optimal protection. Vaccine et al. Rabies virus-based vectors expressing 11359-65 2008;26(50):6329-37 human immunodeficiency virus type 1 167. Schnell MJ, Johnson JE, Buonocore L, 178. Chattopadhyay A, Rose JK. Complementing (HIV-1) envelope protein induce a strong, Rose JK. Construction of a novel virus that defective viruses that express separate cross-reactive cytotoxic T-lymphocyte targets HIV-1-infected cells and controls paramyxovirus glycoproteins provide a new response against envelope proteins from HIV-1 infection. Cell 1997;90(5):849-57 vaccine vector approach. J Virol 2011;85(5): different HIV-1 isolates. J Virol 2001;75(9): 168. Roberts A, Kretzschmar E, Perkins AS, 2004-11 4430-4 et al. Vaccination with a recombinant 179. Lo MK, Bird BH, Chattopadhyay A, et al. 156. Faul EJ, Wanjalla CN, McGettigan JP, vesicular stomatitis virus expressing an Single-dose replication-defective VSV-based Schnell MJ. Interferon-beta expressed by a influenza virus hemagglutinin provides Nipah virus vaccines provide protection rabies virus-based HIV-1 vaccine vector complete protection from influenza virus from lethal challenge in Syrian hamsters. serves as a molecular adjuvant and decreases challenge. J Virol 1998;72(6):4704-11 Antiviral Res 2014;101:26-9 pathogenicity. Virology 2008;382(2):226-38 169. Schwartz JA, Buonocore L, Roberts A, et al. 180. Geisbert TW, Feldmann H. Recombinant 157. McGettigan JP, Koser ML, McKenna PM, Vesicular stomatitis virus vectors expressing vesicular stomatitis virus-based vaccines et al. Enhanced humoral HIV-1-specific avian influenza H5 HA induce against Ebola and Marburg virus infections. immune responses generated from cross-neutralizing antibodies and long-term J Infect Dis 2011;204(Suppl 3):S1075-81 recombinant rhabdoviral-based vaccine protection. Virology 2007;366(1):166-73 •• vectors co-expressing HIV-1 proteins and Reviews the development and preclinical 170. Publicover J, Ramsburg E, Rose JK. IL-2. Virology 2006;344(2):363-77 testing of recombinant vesicular stomatitis A single-cycle vaccine vector based on virus-vectored filovirus vaccines. 158. Faul EJ, Aye PP, Papaneri AB, et al. Rabies vesicular stomatitis virus can induce 181. Chattopadhyay A, Wang E, Seymour R, virus-based vaccines elicit neutralizing immune responses comparable to those et al. A chimeric vesiculo/alphavirus is an antibodies, poly-functional CD8+ T cell, generated by a replication-competent vector. effective alphavirus vaccine. J Virol 2013; and protect rhesus macaques from J Virol 2005;79(21):13231-8 AIDS-like disease after SIV(mac251) 87(1):395-402 171. Cobleigh MA, Buonocore L, Uprichard SL, challenge. Vaccine 2009;28(2):299-308 182. Rose NF, Marx PA, Luckay A, et al. An et al. A vesicular stomatitis virus-based effective AIDS vaccine based on live 159. Gomme EA, Faul EJ, Flomenberg P, et al. hepatitis B virus vaccine vector provides attenuated vesicular stomatitis virus Characterization of a single-cycle rabies protection against challenge in a single dose. recombinants. Cell 2001;106(5):539-49 virus-based vaccine vector. J Virol 2010; J Virol 2010;84(15):7513-22 84(6):2820-31 183. Haglund K, Leiner I, Kerksiek K, et al. 172. Schlereth B, Rose JK, Buonocore L, et al. High-level primary CD8(+) T-cell response 160. Osakada F, Mori T, Cetin AH, et al. New Successful vaccine-induced seroconversion to human immunodeficiency virus rabies virus variants for monitoring and by single-dose immunization in the presence manipulating activity and gene expression in type 1 gag and env generated by vaccination For personal use only. of measles virus-specific maternal antibodies. with recombinant vesicular stomatitis defined neural circuits. Neuron 2011;71(4): J Virol 2000;74(10):4652-7 617-31 viruses. J Virol 2002;76(6):2730-8 173. Kapadia SU, Rose JK, Lamirande E, et al. 184. Wu K, Kim GN, Kang CY. Expression and 161. Roberts A, Buonocore L, Price R, et al. Long-term protection from SARS processing of human immunodeficiency Attenuated vesicular stomatitis viruses as coronavirus infection conferred by a single virus type 1 gp160 using the vesicular vaccine vectors. J Virol 1999;73(5):3723-32 immunization with an attenuated stomatitis virus New Jersey serotype vector 162. Lichty BD, Power AT, Stojdl DF, Bell JC. VSV-based vaccine. Virology 2005;340(2): system. J Gen Virol 2009;90(Pt 5):1135-40 Vesicular stomatitis virus: re-inventing the 174-82 185. Marthas ML, Van Rompay KK, Abbott Z, bullet. Trends Mol Med 2004;10(5):210-16 174. Kahn JS, Schnell MJ, Buonocore L, et al. Partial efficacy of a VSV-SIV/MVA- 163. Bukreyev A, Skiadopoulos MH, Rose JK. Recombinant vesicular stomatitis SIV vaccine regimen against oral SIV Murphy BR, Collins PL. Nonsegmented virus expressing respiratory syncytial virus challenge in infant macaques. Vaccine 2011; negative-strand viruses as vaccine vectors. (RSV) glycoproteins: RSV fusion protein

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 29(17):3124-37 J Virol 2006;80(21):10293-306 can mediate infection and cell fusion. 186. Van Rompay KK. The use of nonhuman 164. Johnson JE, Schnell MJ, Buonocore L, Virology 1999;254(1):81-91 models of HIV infection for the Rose JK. Specific targeting to CD4+ cells of 175. Brown KS, Safronetz D, Marzi A, et al. evaluation of antiviral strategies. AIDS Res recombinant vesicular stomatitis viruses Vesicular stomatitis virus-based vaccine Hum Retroviruses 2012;28(1):16-35 encoding human immunodeficiency virus protects hamsters against lethal challenge envelope proteins. J Virol 1997;71(7): with Andes virus. J Virol 2011;85(23): 187. Lawrence TM, Wanjalla CN, Gomme EA, 5060-8 12781-91 et al. Comparison of Heterologous Prime-Boost Strategies against Human 165. Kretzschmar E, Buonocore L, Schnell MJ, 176. Majid AM, Ezelle H, Shah S, Barber GN. Immunodeficiency Virus Type 1 Gag Using Rose JK. High-efficiency incorporation of Evaluating replication-defective vesicular Negative Stranded RNA Viruses. PLoS functional influenza virus glycoproteins into stomatitis virus as a vaccine vehicle. J Virol ONE 2013;8(6):e67123 recombinant vesicular stomatitis viruses. 2006;80(14):6993-7008 188. Fuchs J, Frank I, Kochar M, et al. J Virol 1997;71(8):5982-9 177. Chattopadhyay A, Park S, Delmas G, et al. First-in-human phase I clinical trial of a 166. Schnell MJ, Buonocore L, Kretzschmar E, Single-dose, virus-vectored vaccine recombinant vesicular stomatitis virus et al. Foreign glycoproteins expressed from protection against Yersinia pestis challenge: (rVSV)-based preventive HIV-1 vaccine. recombinant vesicular stomatitis viruses are CD4+ cells are required at the time of Retrovirology 2012;9(Suppl 2):P134 incorporated efficiently into virus particles.

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

189. Therapeutic Vaccine for HIV. Available type 2 virus from cDNA clones and (BPIV3) fusion and from: http://clinicaltrials.gov/show/ properties of the defective virus without hemagglutinin-neuraminidase glycoproteins NCT01859325 V-specific cysteine-rich domain. Virology make an important contribution to the 190. Gunther S, Feldmann H, Geisbert TW, 2001;284(1):99-112 restricted replication of BPIV3 in primates. et al. Management of accidental exposure to 204. McAuliffe JM, Surman SR, Newman JT, J Virol 2000;74(19):8922-9 Ebola virus in the et al. Codon substitution mutations at two 214. Pennathur S, Haller AA, MacPhail M, et al. 4 laboratory, Hamburg, Germany. J Infect positions in the L polymerase protein of Evaluation of attenuation, immunogenicity Dis 2011;204(Suppl 3):S785-90 human parainfluenza virus type 1 yield and efficacy of a bovine parainfluenza virus 191. Safety and immunogenicity of prime-boost viruses with a spectrum of attenuation type 3 (PIV-3) vaccine and a recombinant vsv ebola vaccine in healthy adults. Available in vivo and increased phenotypic stability in chimeric bovine/human PIV-3 vaccine from: http://apps.who.int/trialsearch/Trial2. vitro. J Virol 2004;78(4):2029-36 vector in rhesus monkeys. J Gen Virol aspx?TrialID=NCT02257840 205. Tao T, Durbin AP, Whitehead SS, et al. 2003;84(Pt 12):3253-61 192. Hastie E, Grdzelishvili VZ. Vesicular Recovery of a fully viable chimeric human 215. Schmidt AC, McAuliffe JM, Murphy BR, stomatitis virus as a flexible platform for parainfluenza virus (PIV) type 3 in which Collins PL. Recombinant bovine/human oncolytic virotherapy against cancer. J Gen the hemagglutinin-neuraminidase and fusion parainfluenza virus type 3 (B/HPIV3) Virol 2012;93(Pt 12):2529-45 glycoproteins have been replaced by those of expressing the respiratory syncytial virus PIV type 1. J Virol 1998;72(4):2955-61 (RSV) G and F proteins can be used to 193. Viral therapy in treating patient with liver achieve simultaneous mucosal immunization cancer. Available from: http://clinicaltrials. 206. Bailly JE, McAuliffe JM, Durbin AP, et al. against RSV and HPIV3. J Virol 2001; gov/show/NCT01628640 A recombinant human parainfluenza virus type 3 (PIV3) in which the nucleocapsid N 75(10):4594-603 194. Nzonza A, Lecollinet S, Chat S, et al. protein has been replaced by that of bovine 216. Schmidt AC, Wenzke DR, McAuliffe JM, A recombinant novirhabdovirus presenting PIV3 is attenuated in primates. J Virol et al. Mucosal immunization of rhesus at the surface the E Glycoprotein from 2000;74(7):3188-95 monkeys against respiratory syncytial virus West Nile Virus (WNV) is immunogenic subgroups A and B and human and provides partial protection against lethal 207. Karron RA, Thumar B, Schappell E, et al. parainfluenza virus type 3 by using a live WNV challenge in BALB/c mice. PLoS Evaluation of two chimeric bovine-human cDNA-derived vaccine based on a host One 2014;9(3):e91766 parainfluenza virus type 3 vaccines in infants and young children. Vaccine 2012;30(26): range-attenuated bovine parainfluenza virus 195. Downham MA, McQuillin J, Gardner PS. 3975-81 type 3 vector backbone. J Virol 2002;76(3): Diagnosis and clinical significance of 1089-99 parainfluenza virus infections in children. 208. van Wyke Coelingh KL, Winter CC, 217. Tang RS, Schickli JH, MacPhail M, et al. Arch Dis Child 1974;49(1):8-15 Tierney EL, et al. Attenuation of bovine parainfluenza virus type 3 in nonhuman Effects of human metapneumovirus and 196. Garbino J, Inoubli S, Mossdorf E, et al. primates and its ability to confer immunity respiratory syncytial virus antigen insertion For personal use only. Respiratory viruses in HIV-infected patients ’ to human parainfluenza virus in two 3 proximal genome positions of with suspected respiratory opportunistic type 3 challenge. J Infect Dis 1988;157(4): bovine/human parainfluenza virus type 3 on infection. Aids 2008;22(6):701-5 655-62 virus replication and immunogenicity. 197. Glezen WP, Frank AL, Taber LH, Kasel JA. J Virol 2003;77(20):10819-28 209. Karron RA, Makhene M, Gay K, et al. Parainfluenza virus type 3: seasonality and Evaluation of a live attenuated bovine 218. Gorman WL, Gill DS, Scroggs RA, risk of infection and reinfection in young parainfluenza type 3 vaccine in two- to Portner A. The children. J Infect Dis 1984;150(6):851-7 six-month-old infants. Pediatr Infect Dis J hemagglutinin-neuraminidase glycoproteins 198. Henrickson KJ. Lower respiratory viral 1996;15(8):650-4 of human parainfluenza virus type 1 and infections in immunocompetent children. Sendai virus have high structure-function 210. Karron RA, Wright PF, Hall SL, et al. Adv Pediatr Infect Dis 1994;9:59-96 similarity with limited antigenic A live attenuated bovine parainfluenza virus cross-reactivity. Virology 1990;175(1): 199. Leader S, Kohlhase K. Respiratory syncytial type 3 vaccine is safe, infectious, 211-21 Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 virus-coded pediatric hospitalizations, immunogenic, and phenotypically stable in 1997 to 1999. Pediatr Infect Dis J 2002; infants and children. J Infect Dis 1995; 219. Lyn D, Gill DS, Scroggs RA, Portner A. 21(7):629-32 171(5):1107-14 The nucleoproteins of human parainfluenza virus type 1 and Sendai virus share amino 200. Le Bayon JC, Lina B, Rosa-Calatrava M, 211. Karron RA, Wright PF, Newman FK, et al. acid sequences and antigenic and structural Boivin G. Recent developments with A live human parainfluenza type 3 virus determinants. J Gen Virol 1991;72(Pt 4): live-attenuated recombinant paramyxovirus vaccine is attenuated and immunogenic in 983-7 vaccines. Rev Med Virol 2013;23(1):15-34 healthy infants and children. J Infect Dis 220. Dave VP, Allan JE, Slobod KS, et al. Viral 201. Durbin AP, Hall SL, Siew JW, et al. 1995;172(6):1445-50 cross-reactivity and antigenic determinants Recovery of infectious human parainfluenza 212. Haller AA, Miller T, Mitiku M, recognized by human parainfluenza virus virus type 3 from cDNA. Virology 1997; Coelingh K. Expression of the surface type 1-specific cytotoxic T-cells. Virology 235(2):323-32 glycoproteins of human parainfluenza virus 1994;199(2):376-83 202. Hoffman MA, Banerjee AK. An infectious type 3 by bovine parainfluenza virus type 3, 221. Hurwitz JL, Soike KF, Sangster MY, et al. clone of human parainfluenza virus type 3. a novel attenuated virus vaccine vector. Intranasal Sendai virus vaccine protects J Virol 1997;71(6):4272-7 J Virol 2000;74(24):11626-35 African green monkeys from infection with 213. Schmidt AC, McAuliffe JM, Huang A, 203. Kawano M, Kaito M, Kozuka Y, et al. human parainfluenza virus-type one. et al. Bovine parainfluenza virus type 3 Recovery of infectious human parainfluenza Vaccine 1997;15(5):533-40

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

222. Smith FS, Portner A, Leggiadro RJ, et al. 234. Chin J, Magoffin RL, Shearer LA, et al. 244. Takeda A, Igarashi H, Nakamura H, et al. Age-related development of human memory Field evaluation of a respiratory syncytial Protective efficacy of an AIDS vaccine, a T-helper and B-cell responses toward virus vaccine and a trivalent parainfluenza single DNA priming followed by a single parainfluenza virus type-1. Virology 1994; virus vaccine in a pediatric population. Am booster with a recombinant 205(2):453-61 J Epidemiol 1969;89(4):449-63 replication-defective Sendai virus vector, in a 223. A Study to assess the safety of live intranasal 235. Fulginiti VA, Eller JJ, Sieber OF, et al. macaque AIDS model. J Virol 2003;77(17): sendai virus vaccine in children and Respiratory virus immunization. I. A field 9710-15 toddlers. Available from: http://clinicaltrials. trial of two inactivated respiratory virus 245. Yu S, Feng X, Shu T, et al. Potent specific gov/show/NCT00186927 vaccines; an aqueous trivalent parainfluenza immune responses induced by 224. Li HO, Zhu YF, Asakawa M, et al. virus vaccine and an alum-precipitated prime-boost-boost strategies based on DNA, A cytoplasmic RNA vector derived from respiratory syncytial virus vaccine. Am J adenovirus, and Sendai virus vectors nontransmissible Sendai virus with efficient Epidemiol 1969;89(4):435-48 expressing gag gene of Chinese gene transfer and expression. J Virol 2000; 236. Mason JN, Elbahesh H, Russell CJ. HIV-1 subtype B. Vaccine 2008;26(48): 74(14):6564-9 Influence of antigen insertion site and vector 6124-31 225. Ferrari S, Griesenbach U, Shiraki-Iida T, dose on immunogenicity and protective 246. Zhang X, Sobue T, Isshiki M, et al. et al. A defective nontransmissible capacity in Sendai virus-based human Elicitation of both anti HIV-1 Env humoral recombinant Sendai virus mediates efficient parainfluenza virus type 3 vaccines. J Virol and cellular immunities by replicating gene transfer to airway epithelium in vivo. 2013;87(10):5959-69 vaccinia prime Sendai virus boost regimen Gene Ther 2004;11(22):1659-64 237. Zhan X, Slobod KS, Krishnamurthy S, and boosting by CD40Lm. PLoS One 2012;7(12):e51633 226. Yonemitsu Y, Kitson C, Ferrari S, et al. et al. Sendai virus recombinant vaccine Efficient gene transfer to airway epithelium expressing hPIV-3 HN or F elicits 247. McEnery R. First candidate HIV vaccine to using recombinant Sendai virus. Nat protective immunity and combines with a employ Sendai vector poised for trials. IAVI Biotechnol 2000;18(9):970-3 second recombinant to prevent hPIV-1, Rep 2013;17(1):18 hPIV-3 and RSV infections. Vaccine 2008; • 227. Hasan MK, Kato A, Shioda T, et al. The first candidate HIV vaccine to 26(27-28):3480-8 Creation of an infectious recombinant employ Sendai vector to enter human Sendai virus expressing the firefly luciferase 238. Le TV, Mironova E, Garcin D, clinical evaluation. ’ Compans RW. Induction of gene from the 3 proximal first locus. J Gen 248. Safety and immunogenicity study of sev-g influenza-specific mucosal immunity by an Virol 1997;78(Pt 11):2813-20 (np) hiv vaccine administered intranasally attenuated recombinant Sendai virus. PLoS 228. Jones B, Zhan X, Mishin V, et al. Human and ad35-grin hiv vaccine given One 2011;6(4):e18780 PIV-2 recombinant Sendai virus (rSeV) intramuscularly in prime-boost regimens in elicits durable immunity and combines with 239. Kano M, Matano T, Kato A, et al. Primary hiv-uninfected volunteers. Available from: replication of a recombinant Sendai virus For personal use only. two additional rSeVs to protect against http://clinicaltrials.gov/show/NCT01705990 vector in macaques. J Gen Virol 2002; hPIV-1, hPIV-2, hPIV-3, and RSV. Vaccine 249. Yonemitsu Y, Matsumoto T, Itoh H, et al. 83(Pt 6):1377-86 2009;27(12):1848-57 DVC1-0101 to treat peripheral arterial 229. Jones BG, Sealy RE, Rudraraju R, et al. 240. Kano M, Matano T, Nakamura H, et al. disease: a Phase I/IIa open-label dose- Sendai virus-based RSV vaccine protects Elicitation of protective immunity against escalation clinical trial. Mol Ther 2013; African green monkeys from RSV infection. simian immunodeficiency virus infection by 21(3):707-14 a recombinant Sendai virus expressing the Vaccine 2012;30(5):959-68 250. Hara H, Hara H, Hironaka T, et al. Gag protein. Aids 2000;14(9):1281-2 230. Jones BG, Sealy RE, Surman SL, et al. Prevalence of specific neutralizing antibodies Sendai virus-based RSV vaccine protects 241. Kawada M, Tsukamoto T, Yamamoto H, against Sendai virus in populations from against RSV challenge in an in vivo et al. Gag-specific cytotoxic different geographic areas: implications for maternal antibody model. Vaccine 2014; T-lymphocyte-based control of primary AIDS vaccine development using Sendai 32(26):3264-73 simian immunodeficiency virus replication virus vectors. Hum Vaccin 2011;7(6): Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 in a vaccine trial. J Virol 2008;82(20): 639-45 231. Takimoto T, Hurwitz JL, Coleclough C, 10199-206 et al. Recombinant Sendai virus expressing 251. Hilleman MR. Current overview of the the G glycoprotein of respiratory syncytial 242. Matano T, Kano M, Nakamura H, et al. pathogenesis and prophylaxis of measles virus (RSV) elicits immune protection Rapid appearance of secondary immune with focus on practical implications. against RSV. J Virol 2004;78(11):6043-7 responses and protection from acute Vaccine 2001;20(5-6):651-65 CD4 depletion after a highly pathogenic 232. Takimoto T, Hurwitz JL, Zhan X, et al. 252. Schwarz AJ. Preliminary tests of a highly immunodeficiency virus challenge in Recombinant Sendai virus as a novel vaccine attenuated measles vaccine. Am J Dis Child macaques vaccinated with a DNA prime/ candidate for respiratory syncytial virus. 1962;103:386-9 Sendai virus vector boost regimen. J Virol Viral Immunol 2005;18(2):255-66 2001;75(23):11891-6 253. Combredet C, Labrousse V, Mollet L, et al. 233. Zhan X, Hurwitz JL, Krishnamurthy S, A molecularly cloned Schwarz strain of 243. Takeda A, Igarashi H, Kawada M, et al. et al. Respiratory syncytial virus (RSV) measles virus vaccine induces strong Evaluation of the immunogenicity of fusion protein expressed by recombinant immune responses in macaques and replication-competent V-knocked-out and Sendai virus elicits B-cell and T-cell transgenic mice. J Virol 2003;77(21): replication-defective F-deleted Sendai virus responses in cotton rats and confers 11546-54 vector-based vaccines in macaques. Vaccine protection against RSV subtypes A and B. 2008;26(52):6839-43 254. Mok H, Cheng X, Xu Q, et al. Evaluation Vaccine 2007;25(52):8782-93 of Measles Vaccine Virus as a Vector to

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

Deliver Respiratory Syncytial Virus Fusion in vitro, and an insight into mechanism(s) implications. Eur J Endocrinol 2006;155(4): Protein or Epstein-Barr Virus Glycoprotein of resistance. Clin Exp Immunol 1998; 495-512 gp350. The open virology journal 2012;6: 113(2):173-82 276. Carlson SK, Classic KL, Hadac EM, et al. 12-22 265. Heinzerling L, Kunzi V, Oberholzer PA, In vivo quantitation of intratumoral 255. Dorig RE, Marcil A, Chopra A, et al. Oncolytic measles virus in cutaneous radioisotope uptake using micro-single Richardson CD. The human T-cell lymphomas mounts antitumor photon emission computed tomography/ CD46 molecule is a receptor for measles immune responses in vivo and targets computed tomography. Mol Imaging Biol virus (Edmonston strain). Cell 1993;75(2): interferon-resistant tumor cells. Blood 2005; 2006;8(6):324-32 295-305 106(7):2287-94 277. Dingli D, Kemp BJ, O’Connor MK, et al. 256. Naniche D, Varior-Krishnan G, Cervoni F, 266. Peng KW, TenEyck CJ, Galanis E, et al. Combined I-124 positron emission et al. Human membrane cofactor protein Intraperitoneal therapy of ovarian cancer tomography/computed tomography imaging (CD46) acts as a cellular receptor for using an engineered measles virus. Cancer of NIS gene expression in animal models of measles virus. J Virol 1993;67(10):6025-32 Res 2002;62(16):4656-62 stably transfected and intravenously 257. Blok VT, Daha MR, Tijsma OM, et al. 267. Peng KW, Hadac EM, Anderson BD, et al. transfected tumor. Mol Imaging Biol 2006; A possible role of CD46 for the protection Pharmacokinetics of oncolytic measles 8(1):16-23 in vivo of human renal tumor cells from virotherapy: eventual equilibrium between 278. Blechacz B, Splinter PL, Greiner S, et al. complement-mediated damage. Lab Invest virus and tumor in an ovarian cancer Engineered measles virus as a novel 2000;80(3):335-44 xenograft model. Cancer Gene Ther 2006; oncolytic viral therapy system for 258. Durrant LG, Spendlove I. Immunization 13(8):732-8 hepatocellular carcinoma. Hepatology 2006; against tumor cell surface 268. Galanis E, Hartmann LC, Cliby WA, et al. 44(6):1465-77 complement-regulatory proteins. Curr Opin Phase I trial of intraperitoneal 279. Dingli D, Diaz RM, Bergert ER, et al. Investig Drugs 2001;2(7):959-66 administration of an oncolytic measles virus Genetically targeted radiotherapy for 259. Fishelson Z, Donin N, Zell S, et al. strain engineered to express multiple myeloma. Blood 2003;102(2): Obstacles to cancer immunotherapy: carcinoembryonic antigen for recurrent 489-96 expression of membrane complement ovarian cancer. Cancer Res 2010;70(3): 280. A Phase II trial of oncolytic virotherapy by regulatory proteins (mCRPs) in tumors. 875-82 systemic administration of edmonston strain Mol Immunol 2003;40(2-4):109-23 269. Hasegawa K, Pham L, O’Connor MK, of measles virus. Available from: http:// 260. Hara T, Suzuki Y, Semba T, et al. High et al. Dual therapy of ovarian cancer using clinicaltrials.gov/show/NCT02192775 expression of membrane cofactor protein of measles viruses expressing carcinoembryonic 281. MV-NIS infected mesenchymal stem cells in complement (CD46) in human leukaemia antigen and sodium iodide symporter. Clin treating patients with recurrent ovarian cell lines: implication of an alternatively Cancer Res 2006;12(6):1868-75 cancer. Available from: http://clinicaltrials.

For personal use only. spliced form containing the STA domain in 270. Peng KW, Facteau S, Wegman T, et al. gov/show/NCT02068794 CD46 up-regulation. Scand J Immunol Non-invasive in vivo monitoring of 282. Intrapleural measles virus therapy in patients 1995;42(6):581-90 trackable viruses expressing soluble marker with malignant pleural mesothelioma. 261. Ong HT, Timm MM, Greipp PR, et al. peptides. Nat Med 2002;8(5):527-31 Available from: http://clinicaltrials.gov/show/ Oncolytic measles virus targets high 271. Phuong LK, Allen C, Peng KW, et al. Use NCT01503177 CD46 expression on multiple myeloma of a vaccine strain of measles virus 283. Viral therapy in treating patients with cells. Exp Hematol 2006;34(6):713-20 genetically engineered to produce recurrent or metastatic squamous cell 262. Seya T, Hara T, Matsumoto M, Akedo H. carcinoembryonic antigen as a novel carcinoma of the head and neck cancer. Quantitative analysis of membrane cofactor therapeutic agent against glioblastoma Available from: http://clinicaltrials.gov/show/ protein (MCP) of complement. High multiforme. Cancer Res 2003;63(10): NCT01846091 2462-9 expression of MCP on human leukemia cell 284. Vaccine therapy with or without lines, which is down-regulated during cell 272. Viral therapy in treating patients with cyclophosphamide in treating patients with Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 differentiation. J Immunol 1990;145(1): recurrent glioblastoma multiforme. Available recurrent or refractory multiple myeloma. 238-45 from: http://clinicaltrials.gov/show/ Available from: http://clinicaltrials.gov/show/ 263. Simpson KL, Jones A, Norman S, NCT00390299 NCT00450814%20MC038C% Holmes CH. Expression of the complement 273. Dingli D, Peng KW, Harvey ME, et al. 20P30CA015083%20MC038C%2006- regulatory proteins decay accelerating factor Image-guided radiovirotherapy for multiple 005263%20NCI-2009-01194% (DAF, CD55), membrane cofactor protein myeloma using a recombinant measles virus 20NCT00450814 (MCP, CD46) and CD59 in the normal expressing the thyroidal sodium iodide 285. Recombinant measles virus vaccine therapy human uterine cervix and in premalignant symporter. Blood 2004;103(5):1641-6 and oncolytic virus therapy in treating and malignant cervical disease. Am J Pathol 274. Mazzaferri EL, Kloos RT. Clinical review patients with progressive, recurrent, or 1997;151(5):1455-67 128: current approaches to primary therapy refractory ovarian epithelial cancer or 264. Varsano S, Rashkovsky L, Shapiro H, et al. for papillary and follicular thyroid cancer. J primary peritoneal cancer. Available from: Human lung cancer cell lines express cell Clin Endocrinol Metab 2001;86(4):1447-63 http://clinicaltrials.gov/show/NCT00408590 membrane complement inhibitory proteins 275. Riesco-Eizaguirre G, Santisteban P. 286. Guerbois M, Moris A, Combredet C, et al. and are extremely resistant to A perspective view of sodium iodide Live attenuated measles vaccine expressing complement-mediated lysis; a comparison symporter research and its clinical HIV-1 Gag virus like particles covered with with normal human respiratory epithelium

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

gp160DeltaV1V2 is strongly immunogenic. 298. Clarke DK, Sidhu MS, Johnson JE, encoding the influenza virus hemagglutinin Virology 2009;388(1):191-203 Udem SA. Rescue of mumps virus from protects against H5N1 highly pathogenic 287. Liniger M, Zuniga A, Morin TN, et al. cDNA. J Virol 2000;74(10):4831-8 avian influenza virus infection following Recombinant measles viruses expressing 299. He B, Paterson RG, Ward CD, Lamb RA. intranasal or intramuscular vaccination of single or multiple antigens of human Recovery of infectious SV5 from cloned BALB/c mice. J Virol 2013;87(1):363-71 immunodeficiency virus (HIV-1) induce DNA and expression of a foreign gene. 310. Phan SI, Chen Z, Xu P, et al. A respiratory cellular and humoral immune responses. Virology 1997;237(2):249-60 syncytial virus (RSV) vaccine based on Vaccine 2009;27(25-26):3299-305 300. Xu R, Nasar F, Megati S, et al. Prime-boost parainfluenza virus 5 (PIV5). Vaccine 2014; 288. Lorin C, Combredet C, Labrousse V, et al. vaccination with recombinant mumps virus 32(25):3050-7 A paediatric vaccination vector based on live and recombinant vesicular stomatitis virus 311. Tompkins SM, Lin Y, Leser GP, et al. attenuated measles vaccine. Therapie 2005; vectors elicits an enhanced human Recombinant parainfluenza virus 5 (PIV5) 60(3):227-33 immunodeficiency virus type 1 Gag-specific expressing the influenza A virus 289. Lorin C, Delebecque F, Labrousse V, et al. cellular immune response in rhesus hemagglutinin provides immunity in mice A recombinant live attenuated measles macaques. J Virol 2009;83(19):9813-23 to influenza A virus challenge. Virology vaccine vector primes effective 301. Parks GD, Alexander-Miller MA. High 2007;362(1):139-50 HLA-A0201-restricted cytotoxic T avidity cytotoxic T lymphocytes to a foreign 312. van den Hoogen BG, de Jong JC, Groen J, lymphocytes and broadly neutralizing antigen are efficiently activated following et al. A newly discovered human antibodies against HIV-1 conserved immunization with a recombinant pneumovirus isolated from young children epitopes. Vaccine 2005;23(36):4463-72 paramyxovirus, simian virus 5. J Gen Virol with respiratory tract disease. Nat Med 290. Lorin C, Mollet L, Delebecque F, et al. 2002;83(Pt 5):116772 2001;7(6):719-24 A single injection of recombinant measles 302. Arimilli S, Johnson JB, Clark KM, et al. 313. Hamelin ME, Abed Y, Boivin G. Human virus vaccines expressing human Engineered expression of the TLR5 ligand metapneumovirus: a new player among immunodeficiency virus (HIV) type 1 clade flagellin enhances paramyxovirus activation respiratory viruses. Clin Infect Dis 2004; B envelope glycoproteins induces of human dendritic cell function. J Virol 38(7):983-90 neutralizing antibodies and cellular immune 2008;82(22):10975-85 314. Biacchesi S, Skiadopoulos MH, Tran KC, responses to HIV. J Virol 2004;78(1): 303. Chen Z, Xu P, Salyards GW, et al. et al. Recovery of human metapneumovirus 146-57 Evaluating a parainfluenza virus 5-based from cDNA: optimization of growth 291. Lorin C, Segal L, Mols J, et al. Toxicology, vaccine in a host with pre-existing in vitro and expression of additional genes. biodistribution and shedding profile of a immunity against parainfluenza virus 5. Virology 2004;321(2):247-59 recombinant measles vaccine vector PLoS One 2012;7(11):e50144 315. Govindarajan D, Buchholz UJ, Samal SK. expressing HIV-1 antigens, in cynomolgus 304. Chen Z, Zhou M, Gao X, et al. A novel Recovery of avian metapneumovirus macaques. Naunyn Schmiedebergs Arch For personal use only. rabies vaccine based on a recombinant subgroup C from cDNA: cross-recognition Pharmacol 2012;385(12):1211-25 parainfluenza virus 5 expressing rabies virus of avian and human metapneumovirus 292. Stebbings R, Fevrier M, Li B, et al. glycoprotein. J Virol 2013;87(6):2986-93 support proteins. J Virol 2006;80(12): Immunogenicity of a recombinant 305. Clark KM, Johnson JB, Kock ND, et al. 5790-7 measles-HIV-1 clade B candidate vaccine. Parainfluenza virus 5-based vaccine vectors 316. Biacchesi S, Skiadopoulos MH, Yang L, PLoS One 2012;7(11):e50397 expressing vaccinia virus (VACV) antigens et al. Recombinant human 293. Stebbings R, Li B, Lorin C, et al. provide long-term protection in mice from Metapneumovirus lacking the small Immunogenicity of a recombinant measles lethal intranasal VACV challenge. Virology hydrophobic SH and/or attachment G HIV-1 subtype C vaccine. Vaccine 2013; 2011;419(2):97-106 glycoprotein: deletion of G yields a 31(51):6079-86 306. Li Z, Gabbard JD, Mooney A, et al. promising vaccine candidate. J Virol 2004; 294. Wang Z, Hangartner L, Cornu TI, et al. Efficacy of parainfluenza virus 5 mutants 78(23):12877-87 Recombinant measles viruses expressing expressing hemagglutinin from 317. Buchholz UJ, Biacchesi S, Pham QN, et al. Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 heterologous antigens of mumps and simian H5N1 influenza A virus in mice. J Virol Deletion of M2 gene open reading frames immunodeficiency viruses. Vaccine 2001; 2013;87(17):9604-9 1 and 2 of human metapneumovirus: effects 19(17-19):2329-36 307. Li Z, Gabbard JD, Mooney A, et al. on RNA synthesis, attenuation, and 295. Zuniga A, Liniger M, Morin TN, et al. Single-dose vaccination of a recombinant immunogenicity. J Virol 2005;79(11): Sequence and immunogenicity of a clinically parainfluenza virus 5 expressing NP from 6588-97 approved novel measles virus vaccine vector. H5N1 virus provides broad immunity 318. Biacchesi S, Pham QN, Skiadopoulos MH, Hum Vaccin Immunother 2013;9(3): against influenza A viruses. J Virol 2013; et al. Infection of nonhuman primates with 607-13 87(10):5985-93 recombinant human metapneumovirus 296. Zuniga A, Wang Z, Liniger M, et al. 308. Li Z, Mooney AJ, Gabbard JD, et al. lacking the SH, G, or M2-2 protein Attenuated measles virus as a vaccine vector. Recombinant parainfluenza virus categorizes each as a nonessential accessory Vaccine 2007;25(16):2974-83 5 expressing hemagglutinin of influenza protein and identifies vaccine candidates. J Virol 2005;79(19):12608-13 297. Study to evaluate the dosage and safety of A virus H5N1 protected mice against lethal two intramuscular injections of an highly pathogenic avian influenza virus 319. Pham QN, Biacchesi S, Skiadopoulos MH, investigational clade B HIV vaccine. H5N1 challenge. J Virol 2013;87(1):354-62 et al. Chimeric recombinant human Available from: http://clinicaltrials.gov/show/ 309. Mooney AJ, Li Z, Gabbard JD, et al. metapneumoviruses with the nucleoprotein NCT01320176 Recombinant parainfluenza virus 5 vaccine or phosphoprotein open reading frame

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines RNA-based viral vectors Review

replaced by that of avian metapneumovirus 330. Goff PH, Krammer F, Hai R, et al. 340. Blodorn K, Hagglund S, Fix J, et al. exhibit improved growth in vitro and Induction of cross-reactive antibodies to Vaccine safety and efficacy evaluation of a attenuation in vivo. J Virol 2005;79(24): novel H7N9 influenza virus by recombinant recombinant bovine respiratory syncytial 15114-22 Newcastle disease virus expressing a North virus (BRSV) with deletion of the SH gene 320. Evaluation of the safety and American lineage H7 subtype and subunit vaccines based on recombinant immunogenicity of a live attenuated human hemagglutinin. J Virol 2013;87(14):8235-40 human RSV proteins: N-nanorings, P and metapneumovirus Vaccine. Available from: 331. Nakaya T, Cros J, Park MS, et al. M2-1, in calves with maternal antibodies. http://clinicaltrials.gov/show/NCT01255410 Recombinant Newcastle disease virus as a PLoS ONE 2014;9(6):e100392 321. Alexander DJ. Newcastle disease and other vaccine vector. J Virol 2001;75(23): 341. Karron RA, Wright PF, Belshe RB, et al. avian paramyxovirus infections. In: Diseases 11868-73 Identification of a recombinant live of poultry. Calnek BW, Barnes HJ, 332. DiNapoli JM, Yang L, Samal SK, et al. attenuated respiratory syncytial virus vaccine Beard CW, Reid WM and Yoder HW, Respiratory tract immunization of candidate that is highly attenuated in Editors Iowa State University Press; Ames, non-human primates with a Newcastle infants. J Infect Dis 2005;191(7):1093-104 IA: 1991. p. 496-519 disease virus-vectored vaccine candidate 342. Taylor G, Wyld S, Valarcher JF, et al. 322. Peeters BP, de Leeuw OS, Koch G, against Ebola virus elicits a neutralizing Recombinant bovine respiratory syncytial Gielkens AL. Rescue of Newcastle disease antibody response. Vaccine 2010;29(1): virus with deletion of the SH gene induces virus from cloned cDNA: evidence that 17-25 increased apoptosis and pro-inflammatory cleavability of the fusion protein is a major 333. DiNapoli JM, Yang L, Suguitan A Jr, et al. cytokines in vitro, and is attenuated and determinant for virulence. J Virol 1999; Immunization of primates with a Newcastle induces protective immunity in calves. J 73(6):5001-9 disease virus-vectored vaccine via the Gen Virol 2014;95(Pt 6):1244-54 323. Romer-Oberdorfer A, Mundt E, respiratory tract induces a high titer of 343. Kwilas AR, Yednak MA, Zhang L, et al. Mebatsion T, et al. Generation of serum neutralizing antibodies against highly Respiratory syncytial virus engineered to recombinant lentogenic Newcastle disease pathogenic avian influenza virus. J Virol express the cystic fibrosis transmembrane virus from cDNA. J Gen Virol 1999; 2007;81(21):11560-8 conductance regulator corrects the 80(Pt 11):2987-95 334. DiNapoli JM, Ward JM, Cheng L, et al. bioelectric phenotype of human cystic fibrosis airway epithelium in vitro. J Virol 324. Fournier P, Schirrmacher V. Oncolytic Delivery to the lower respiratory tract is 2010;84(15):7770-81 Newcastle Disease Virus as Cutting Edge required for effective immunization with between Tumor and Host. Biology 2013; Newcastle disease virus-vectored vaccines 344. Malykhina O, Yednak MA, Collins PL, 2(3):936-75 intended for humans. Vaccine 2009;27(10): et al. A respiratory syncytial virus replicon 1530-9 that is noncytotoxic and capable of 325. Park MS, Steel J, Garcia-Sastre A, et al. long-term foreign gene expression. J Virol Engineered viral vaccine constructs with 335. Martinez-Sobrido L, Gitiban N, Fernandez-Sesma A, et al. Protection against 2011;85(10):4792-801 For personal use only. dual specificity: avian influenza and Newcastle disease. Proc Natl Acad Sci USA respiratory syncytial virus by a recombinant 345. Brand D, Lemiale F, Turbica I, et al. 2006;103(21):8203-8 Newcastle disease virus vector. J Virol 2006; Comparative analysis of humoral immune 80(3):1130-9 responses to HIV type 1 envelope 326. Veits J, Wiesner D, Fuchs W, et al. glycoproteins in mice immunized with a Newcastle disease virus expressing 336. Carnero E, Li W, Borderia AV, et al. DNA vaccine, recombinant Semliki Forest H5 hemagglutinin gene protects chickens Optimization of human immunodeficiency virus RNA, or recombinant Semliki Forest against Newcastle disease and avian virus gag expression by newcastle disease virus particles. AIDS Res Hum Retroviruses influenza. Proc Natl Acad Sci USA 2006; virus vectors for the induction of potent 1998;14(15):1369-77 103(21):8197-202 immune responses. J Virol 2009;83(2): 584-97 346. Fleeton MN, Sheahan BJ, Gould EA, et al. 327. DiNapoli JM, Kotelkin A, Yang L, et al. Recombinant Semliki Forest virus particles Newcastle disease virus, a host 337. Maamary J, Array F, Gao Q, et al. encoding the prME or NS1 proteins of range-restricted virus, as a vaccine vector for Newcastle disease virus expressing a

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 louping ill virus protect mice from lethal intranasal immunization against emerging dendritic cell-targeted HIV gag protein challenge. J Gen Virol 1999;80(Pt 5): pathogens. Proc Natl Acad Sci USA 2007; induces a potent gag-specific immune 1189-98 104(23):9788-93 response in mice. J Virol 2011;85(5): 2235-46 347. Berglund P, Fleeton MN, Smerdou C, 328. DiNapoli JM, Nayak B, Yang L, et al. Liljestrom P. Immunization with Newcastle disease virus-vectored vaccines 338. Jin H, Clarke D, Zhou HZ, et al. recombinant Semliki Forest virus induces expressing the hemagglutinin or Recombinant human respiratory syncytial protection against influenza challenge in neuraminidase protein of H5N1 highly virus (RSV) from cDNA and construction mice. Vaccine 1999;17(5):497-507 pathogenic avian influenza virus protect of subgroup A and B chimeric RSV. against virus challenge in monkeys. J Virol Virology 1998;251(1):206-14 348. Daemen T, Pries F, Bungener L, et al. 2010;84(3):1489-503 339. Collins PL. The molecular biology of Genetic immunization against cervical carcinoma: induction of cytotoxic T 329. Ge J, Deng G, Wen Z, et al. Newcastle human respiratory syncytial virus (RSV) of lymphocyte activity with a recombinant disease virus-based live attenuated vaccine genus Pneumovirus. In: The alphavirus vector expressing human completely protects chickens and mice from Paramyxoviruses. David WK, Editor papillomavirus type 16 E6 and E7. Gene lethal challenge of homologous and Plenum Publishing Corp; New york: 1991. Ther 2000;7(21):1859-66 heterologous H5N1 avian influenza viruses. p. 103-62 J Virol 2007;81(1):150-8 349. Vidalin O, Fournillier A, Renard N, et al. Use of conventional or replicating nucleic

informahealthcare.com doi: 10.1586/14760584.2015.979798 Review Mogler & Kamrud

acid-based vaccines and recombinant 358. Loy JD, Gander J, Mogler M, et al. 367. Bukreyev A, Marzi A, Feldmann F, et al. Semliki forest virus-derived particles for the Development and evaluation of a replicon Chimeric human parainfluenza virus bearing induction of immune responses against particle vaccine expressing the the Ebola virus glycoprotein as the sole hepatitis C virus core and E2 antigens. E2 glycoprotein of bovine viral diarrhea surface protein is immunogenic and highly Virology 2000;276(2):259-70 virus (BVDV) in cattle. Virol J 2013;10:35 protective against Ebola virus challenge. 350. Nilsson C, Makitalo B, Berglund P, et al. 359. Vander Veen RL, Mogler MA, Russell BJ, Virology 2009;383(2):348-61 Enhanced simian immunodeficiency et al. Haemagglutinin and nucleoprotein 368. Skiadopoulos MH, Biacchesi S, virus-specific immune responses in replicon particle vaccination of swine Buchholz UJ, et al. Individual contributions macaques induced by priming with protects against the pandemic H1N1 of the human metapneumovirus F, G, and recombinant Semliki Forest virus and 2009 virus. The Veterinary record 2013; SH surface glycoproteins to the induction of boosting with modified vaccinia virus 173(14):344 neutralizing antibodies and protective Ankara. Vaccine 2001;19(25-26):3526-36 360. Harvey TJ, Anraku I, Linedale R, et al. immunity. Virology 2006;345(2):492-501 351. Brinster C, Chen M, Boucreux D, et al. Kunjin virus replicon vectors for human 369. Tang RS, MacPhail M, Schickli JH, et al. Hepatitis C virus non-structural protein immunodeficiency virus vaccine Parainfluenza virus type 3 expressing the 3-specific cellular immune responses development. J Virol 2003;77(14):7796-803 native or soluble fusion (F) Protein of following single or combined immunization 361. Li SH, Li XF, Zhao H, et al. Development Respiratory Syncytial Virus (RSV) confers with DNA or recombinant Semliki Forest and characterization of the replicon system protection from RSV infection in African virus particles. J Gen Virol 2002;83(Pt 2): of Japanese encephalitis live vaccine virus green monkeys. J Virol 2004;78(20): 369-81 SA14-14-2. Virol J 2013;10:64 11198-207 352. Tsuji M, Bergmann CC, Takita-Sonoda Y, 362. McKenna PM, Koser ML, Carlson KR, 370. Tang RS, Mahmood K, Macphail M, et al. et al. Recombinant Sindbis viruses et al. Highly attenuated rabies virus-based A host-range restricted parainfluenza virus expressing a cytotoxic T-lymphocyte epitope vaccine vectors expressing simian-human type 3 (PIV3) expressing the human of a malaria parasite or of influenza virus immunodeficiency virus89.6P Env and metapneumovirus (hMPV) fusion protein elicit protection against the corresponding simian immunodeficiency virusmac239 Gag elicits protective immunity in African green pathogen in mice. J Virol 1998;72(8): are safe in rhesus macaques and protect monkeys. Vaccine 2005;23(14):1657-67 6907-10 from an AIDS-like disease. J Infect Dis 371. Tang RS, Spaete RR, Thompson MW, 353. Kamrud KI, Hooper JW, Elgh F, 2007;195(7):980-8 et al. Development of a PIV-vectored RSV Schmaljohn CS. Comparison of the 363. Buchholz UJ, Granzow H, Schuldt K, et al. vaccine: preclinical evaluation of safety, protective efficacy of naked DNA, Chimeric bovine respiratory syncytial virus toxicity, and enhanced disease and initial DNA-based Sindbis replicon, and packaged with glycoprotein gene substitutions from clinical testing in healthy adults. Vaccine Sindbis replicon vectors expressing human respiratory syncytial virus (HRSV): 2008;26(50):6373-82 Hantavirus structural genes in hamsters. effects on host range and evaluation as a 372. Bukreyev A, Huang Z, Yang L, et al. For personal use only. Virology 1999;263(1):209-19 live-attenuated HRSV vaccine. J Virol 2000; Recombinant newcastle disease virus 354. Cheng WF, Hung CF, Hsu KF, et al. 74(3):1187-99 expressing a foreign viral antigen is Enhancement of sindbis virus self-replicating 364. Durbin AP, Skiadopoulos MH, attenuated and highly immunogenic in RNA vaccine potency by targeting antigen McAuliffe JM, et al. Human parainfluenza primates. J Virol 2005;79(21):13275-84 to endosomal/lysosomal compartments. virus type 3 (PIV3) expressing the 373. Brandler S, Tangy F. Recombinant vector Hum Gene Ther 2001;12(3):235-52 hemagglutinin protein of measles virus derived from live attenuated measles virus: 355. Heise MT, Whitmore A, Thompson J, provides a potential method for potential for flavivirus vaccines. Comp et al. An alphavirus replicon-derived immunization against measles virus and Immunol Microbiol Infect Dis 2008; candidate vaccine against Rift Valley fever PIV3 in early infancy. J Virol 2000;74(15): 31(2-3):271-91 virus. Epidemiol Infect 2009;137(9): 6821-31 374. Myers R, Harvey M, Kaufmann TJ, et al. 1309-18 365. Skiadopoulos MH, Surman SR, Riggs JM, Toxicology study of repeat intracerebral

Expert Review of Vaccines Downloaded from informahealthcare.com by 174.97.229.189 on 11/08/14 356. Zhu W, Fu J, Lu J, et al. Induction of et al. Evaluation of the replication and administration of a measles virus derivative humoral and cellular immune responses immunogenicity of recombinant human producing carcinoembryonic antigen in against hepatitis C virus by vaccination with parainfluenza virus type 3 vectors expressing rhesus macaques in support of a phase I/II replicon particles derived from Sindbis-like up to three foreign glycoproteins. Virology clinical trial for patients with recurrent virus XJ-160. Arch Virol 2013;158(5): 2002;297(1):136-52 gliomas. Hum Gene Ther 2008;19(7):690-8 1013-19 366. Bukreyev AA, Dinapoli JM, Yang L, et al. 375. Iankov ID, Haralambieva IH, Galanis E. 357. Sashihara J, Hoshino Y, Bowman JJ, et al. Mucosal parainfluenza virus-vectored vaccine Immunogenicity of attenuated measles virus Soluble rhesus against Ebola virus replicates in the engineered to express Helicobacter pylori gp350 protects against infection and reduces respiratory tract of vector-immune monkeys neutrophil-activating protein. Vaccine 2011; viral loads in animals that become infected and is immunogenic. Virology 2010;399(2): 29(8):1710-20 with virus after challenge. PLoS Pathog 290-8 2011;7(10):e1002308

doi: 10.1586/14760584.2015.979798 Expert Rev. Vaccines