New vector and vaccine platforms: mRNA, DNA, viral vectors

INTRODUCTION

In the U.S., there are currently 28 pathogens with a licensed vaccine [1]. Many more are still awaiting an effective vaccine strategy. For more complex diseases such as AIDS, tuberculosis (TB), malaria, cancers, autoimmune and neurodegenerative diseases, vaccine development requires a much greater understanding of the principles and pathways towards effective immunity. Intensive research into HIV-1 vaccines is entering its fifth decade, although with fewer than 10 funded phase 3 trials testing preventive efficacy of candidate vaccines since the start of the AIDS epidemic (compared with nine licensed vaccines and over three dozen vaccine efficacy trials in the first 2 years of the COVID-19 pandemic [2]). Regrettably, the few trialled HIV-1 vaccine strategies have failed to show deployable, reproducible efficacy [3–6]. The lack of efficacy of the candidate vaccines tested to date have been due to product failures [7] rather than to the intrinsic imposibility to elicit actively protective levels of broadly neutralizing antibodies [6] or that robust, broad and well matched killer T cells could not control HIV-1 [8,9]. The devil is in the vaccine details. Conceptually, vaccines have several key components, which together ensure generation of protective responses, and any one of these components performing suboptimally can render the whole vaccine strategy ineffective [10]. These include at least the HIV-1-derived immunogens, their delivery, vaccine formulation and immunomodulation [11▪,12,13]. Although some argue that HIV-1 research is now an immunogen discovery problem, the importance of other components is not to be underestimated.

Vaccines induce host immune responses through a complex set of interactions with multiple cell types, innate immune receptors and pathways, and the mechanisms required for protection vary from pathogen to pathogen. Furthermore, individuals develop different ways of controlling the same virus depending on their genetic makeup [14,15], abundance and molecular state of their preexisting immune cells, including history of other infections offering cross-reactivity and their unique encounter with HIV-1 [16]. The Holy Grail of HIV-1 vaccinology is the induction of broadly neutralizing antibodies. Given the challenges bnAb induction faces and the clear evidence that T cells impose a selective pressure on HIV-1 during natural infection [17], effective vaccines should harness killer T cells if only to complement bnAbs [8,17]. Killer T cells might be particularly important for cure because they are better at killing infected cells than antibodies. In turn, antibodies may enhance T-cell responses through a ‘vaccinal effect’ [18]. Clearly, vaccines must induce better immune responses that natural HIV-1 infection.

In this article, I provide a brief overview of the challenges and opportunities provided by the RNA, DNA and viral vector platforms delivering HIV-1 immunogens. 

FB1Box 1:

no caption available

RAPIDLY EVOLVING LANDSCAPE OF THE RNA VACCINE PLATFORM

Vaccines based on RNA are the ‘hottest’ vaccinology development with the momentum and potential to transform the field [11▪,19–21]. However, RNA alone will not solve all the current vaccine roadblocks for the most difficult diseases, because which immunogens RNA expresses and how it is formulated also matters [22]. Nevertheless, RNA can serve as a tool for rapid iterative optimization of vaccines in humans and significantly accelerate the rate of progress. Intramuscular RNA injection as a strategy for protection against or cure for a variety of cancers and infections including HIV-1 has been in development since late 1980s. Initial obstacles of instability, inefficient delivery, activation of innate immunity through intracellular RNA sensors and large-scale manufacturing have been overcome sufficiently to allow Moderna's mRNA-1273 and BioNTech/Pfizer's BNT162b2 vaccines to emerge during the COVID-19 pandemic as well tolerated and efficacious vaccines [23,24]. At the time of writing this article and as a reflection of the interest in this platform, 37 other RNA-based COVID-19 vaccines are in clinical development representing 22% of the total global COVID-19 vaccine effort [2].

The RNA platform is a type of genetic vaccines. Instead of delivering immunogenic proteins, genetic vaccines provide the protein's coding sequence, which instructs the host cells how to produce the vaccine immunogens. RNA vaccines can be delivered directly as messenger RNA (mRNA) [19] and as a self-replicating RNA (srRNA) [11▪]. mRNA vaccines have smaller molecules and can be used in homologous boosts without reduced efficacy due to antivector immunity. The advantage of srRNA is that after entering cells, it replicates and increases the copy number of the mRNA. This translates into higher immunogen production leading to improved immunogenicity and ultimately lower vaccine doses. srRNAs are purpose engineered replicons not only derived mostly from alphaviruses Venezuelan Equine Encephalitis, Sindbis and Semliki Forest viruses, but also from flaviruses, nodamura viruses and nidoviruses. Although the successful COVID-19 vaccines of Moderna and BioNTech were both nonreplicating mRNAs, srRNA features are attractive and its performance in humans is improving [11▪].

Design of the mRNA nucleotide sequence begins with the decision on the immunogen. This is typically coupled to a strong N-terminal leader sequence, and for HIV-1 Env contains various intramolecular as well as trimer-stabilizing and glycosylation-finetuning amino acid modifications. The ‘know-how’ in the designing of the mRNA molecule itself is nontrivial. The RNA molecule starts with a modified dinucleotide GA (guanine-adenosine), called the 5’ cap, which enhances mRNA stability by impacting on innate sensing. Both the 5’ and 3’-untranslated regions (UTRs) contain critical regulatory elements [21,25,26] and can be derived from highly expressed human genes or use the pathogen's original mRNA UTRs assuming optimization by natural evolution. Translation initiation is often the rate-limiting step in protein synthesis and begins from the first methionine start codon enhanced by a modified Kozak consensus sequence and its flanking secondary RNA structure directing the ribosome subunit to the first AUG. One advantage of natural UTRs is that they are almost certainly devoid of hidden upstream start codons. Taking the publicly available BioNTech/Pfizer SARS-CoV-2 vaccine BNT162b2 sequence [27] as an illustration, the 5’-UTR contains 52 nucleotides, which originate from the highly expressed human α-globin gene. Codons are optimized according to highly expressed human genes, which not only makes them generally GC-rich, but also for the accuracy of translation [28▪▪]. This, together with a higher availability of corresponding amino acid charged transfer RNAs, leads to more efficient protein expression. An interesting safety feature is the reduction of the protein-coding potential by avoiding short overlapping ORFs on both the positive and, for srRNA, negative RNA strands. Notably, mRNA-1273 eliminated all overlapping ORFs on the positive strand compared with eight shorter ORFs found on BNT162b2 and 11 on the wild-type SARS-CoV-2 spike gene [29]. The main ORF ends with two (BNT162b2) or three (mRNA-1273) consecutive stop codons to compensate for pseudouridine (see below) mispairing. The 3’-UTR follows, which is critical for mRNA localization, stability and translational efficacy. The 136 nucleotides of the BNT162b2's 3’-UTR is like the amino-terminal enhancer of split (AES) mRNA improved by computation to minimize miRNA binding. The 3’ end of mRNA is polyadenylated, which further protects against degradation. The optimal length of polyA is approximately 120 A's. The BNT162b2 vaccine ends with 30 A's, a 10-mixed nucleotide spacer and a further 70 A's, whereby the mixed-nucleotide spacer facilitates the manufacturing process involving the template plasmid. Several software assist the mRNA designs by analysing the ORF for codon frequencies, codon adaptation index, index of translation efficiency, position weight matrix and minimum folding energy [28▪▪].

RNA is inherently immunostimulatory to the innate immune system through interaction with pattern-recognition receptors. This leads to the production of type I interferon and pro-inflammatory cytokines. Type I interferon response matures dendritic cells but can be also counterproductive by shutting down the protein synthesis (including that of the immunogen), altering or interfering with protein processing and presentation, affecting autophagy and inducing cellular stress response. All these processes influence the vaccine immunogenicity and reactogenicity. To decrease innate stimulation, RNAs often employ modified nucleosides. Both BNT162b2 and mRNA-1273 replaced uracil with N1-methyl-pseudouridine, but other modifications such 5-methylcytidine can be also used [30,31]. However, the pairing of modified nucleotides is less accurate. Importantly, responses to modified and unmodified mRNA differ between mice, nonhuman primates and humans, thus fine tuning of the vaccine is best done in the relevant species.

Once the mRNA molecule is designed and several mRNAs can compose a single vaccine, it is produced and purified comprehensively from dsRNA and other manufacturing impurities [32]. For efficient delivery, RNA must be protected from degradation by a lipid formulation, which can have a form of liposomes, lipoplex, lipid nanoparticles (LNPs) or polymeric nanoparticles [11▪,19–21]. A typical LNP contains four important excipients: cationic lipids neutralizing RNA's negative charge or a new generation of ionizable amino lipids, which change charge in acidified endosomes to facilitate the RNA dissociation and endosomal exit into cytoplasm, a helper phospholipid supporting the lipid bilayer, cholesterol adjusting the membrane fluidity and polyethylene glycol-lipid improving colloidal stability and shielding from opsonization. The formulation must be of minimum toxicity, which is greatly increased by biodegradable linkages [33]. It also facilitates several important features such as membrane crossing, which may employ cell-penetrating peptides, or specific cell-targeting by attachment of ligands. Cytoplasmic delivery of naked RNA can be increased by electroporation. Every new product will likely need optimization by preparing a few starting RNA variants and formulations to select empirically the most immunogenic blend. BioNtech tested five COVID-19 vaccine candidates to select BNT162b2.

Candidate RNA constructs for HIV-1 were used initially to pulse monocyte-derived dendritic cells, which were transferred back into patients. This strategy showed moderate immunogenicity but offered no or very limited viraemic control [34]. The first HIV-1 mRNA vaccine study was launched in 2015 and was a phase 1 dose-escalation trial in chronically infected patients stably controlling HIV-1 by ART. Naked HIVACAT T-cell immunogen (HTI) [35] mRNA adjuvanted with TriMix mRNAs encoding two dendritic cell activating proteins cluster of differentiation (CD)40L and caTLR4, a constitutively active form of TLR4, as well as T cell-activating CD70 [36] was delivered intranodally and was well tolerated (NCT02413645) [37]. The follow up phase 2a trial showed no immunogenicity, no decrease in latent HIV-1 reservoir and no post-ART control in part due to mRNA design error (NCT02888756) [38]. In November 2021, LNP mRNA vaccines eOD-GT8 60mer (mRNA-1644) and Core-g28v2 60mer mRNA (mRNA-1644v2-Core) self-assembling into virus-like particles entered clinical evaluation and are tested individually and in combination in HIV-1-negative volunteers (NCT05414786). The aim of the eOD-GT8 immunogen is to engage the unmutated germline B-cell precursor with the potential to evolve through somatic mutations into a broadly neutralizing glycan-dependent VRC01-class antibody [39]. IAVI G002 followed in May 2022 delivering eOD-GT8 HIV-1 60mer mRNA to HIV-1 negative African adults (NCT05001373). Finally, in March this year, a phase 1 trial of HIV Vaccine Trials Network (HVTN) 302, opened recruitment administering three variants of HIV-1 Env trimers BG505 MD39.3, BG505 MD39.3 gp151 and BG505 MD39.3 gp151 CD4KO expressed from encapsulated mRNA and given to healthy HIV-1-negative volunteers at multiple sites in the USA (NCT05217641). All the Env LNP mRNA vaccines were produced by Moderna and the results from these studies are expected in 2023. For HIV-1, the landscape of mRNA vaccines will evolve rapidly.

MAJOR ADVANCEMENTS FOR DNA VACCINES ARE STILL AWAITED

DNA began development as a vaccine modality at approximately the same time as RNA [40] and spawned tremendous expectations [41]. The big difference for DNA is that it must cross the cytoplasmic as well as nuclear membranes for expression. The transport of transcribed RNA to cytoplasm for translation can be enhanced by the addition of splice donor and acceptor sites, but caution must be taken not to inadvertently introduce other splicing signals within the 5’-/3’-UTR and ORF design. DNA has been immunogenic in mice and a few other preclinical models, but its immunogenicity in humans remains moderate at best. Up to 8-mg doses, codelivery with cytokines and chemokines [42], biojector devises [43], DNA-coated gold particle intradermal delivery (gene gun) [44], intradermal or intramuscular electroporation [42,45], tattooing [46], magnetoporation/sonoporation/optoporation [47], transcutaneous microneedle strategies [48], dermavir [49], depletion of normal human plasma major DNA-binding protein [50] are either awaiting human testing or have so far failed to deliver a major breakthrough of the DNA platform for human use [7,51]. Moderate improvements by co-delivery of interleukin (IL)-12 and electroporation in trial HVTN 098 (NCT02431767) were observed [52], but these are still easily dwarfed by other delivery modalities. As a prime, DNA increased responses following viral vector boost [45,53–55]. It is interesting whether in this setting, the low immunogen expression following plasmid DNA priming improves the quality and durability of responses after the viral vector boost [45,56]. Currently, there are 16 DNA vaccines in clinical testing against COVID-19 representing 10% of the global COVID-19 vaccine effort [2].

THE POTENTIAL OF VIRAL VECTORS IS IN THEIR DIVERSITY

Development of well tolerated purpose-designed viral vectors is extremely important. There is an unmet need for engineered vaccine vectors and modalities to cover the global demand for safer and more efficient subunit vaccines than those currently in use, against difficult pathogens such as HIV-1, and all the identified and yet unknown pandemic threats. The aim is to develop a panel of well tolerated viruses, which can be administered not only as stand-alone vaccines, but also in heterologous prime-boost regimens to complement and enhance each other, provide sufficient options to rescue first-line vaccine nonresponders and ensure efficient long-term maintenance of immunity by alternating heterologous boosts [8]. Vaccine vectors most commonly under development include those derived from human adenoviruses of serotypes 4, 5, 26 and 35 [4,57,58], simian adenoviruses isolated from chimpanzees [8,59] and gorillas [60,61], vesicular stomatitis virus (VSV) [62], adeno-associated viruses (AAV) [63], integration-deficient lentiviruses (IDLV) [64,65], poxviruses such as vaccinia virus, modified vaccinia Ankara (MVA), fowl poxvirus and human cytomegalovirus (HCMV) to name a few. Vector platforms have been comprehensively reviewed many times over, most recently in a well illustrated review by Travieso et al.[66]. The first viral vector that was given Emergency Use Authorization and the most widely globally used vaccine against SARS-CoV-2 is vectored by ChAdOx1, which was engineered from a simian adenovirus Y25 isolated from a chimpanzee [67]. It is replication deficient, whereby for the manufacture, the essential missing function must be supplied in trans by a stably transformed cell line [68]. These cells also produce a tetracycline repressor slowing substantially the transgene transcription and therefore lowering the biological burden on the vaccine-producing cells during manufacture; this in turn increases vaccine yield and stability. For transgenes, care must be taken not to introduce unwanted splicing events, which can produce reactogenic products. Splicing prediction is inherently unreliable; thus, experimental testing may become a standard [69▪▪,70]. ChAdOx1 is explored by two candidate HIV-1 vaccines delivering conserved mosaic (HIVconsvX) [59] (NCT04586673 and NCT04553016) and protective (HTI) [35] (NCT03204617) T-cell regions. Nonreplicating and replicating virus vectors represent 21 (13%) and 4 (2%) of the current anti-SARS-CoV-2 vaccine strategies in clinical testing, respectively [2].

Virus vectors come in many different ‘shapes and forms’. Vectors can carry single or double stranded nucleic acids, RNA or DNA, a single positive or negative RNA strand, be enveloped or nonenveloped and simple or complex (expressing from a few to over 200 viral proteins). Some vectors can replicate in the host, and others are replication deficient. Replication makes vaccines more immunogenic but requires extra safety vigilance. Vector tropism is dictated by the ligands for cell entry receptors viruses employ. Specific surface markers can be explored for targeting vaccines to professional antigen-presenting cells for efficient induction of immune responses [65]. Viral RNA, DNA and also other pathogen-associated molecular patterns intrinsic to vectors are a powerful stimulus to the innate immune system. Like RNA vaccines, innate pathways can enhance immunogenicity, but also impact on protein expression and presentation and bias the stimulation of adaptive immune responses. Hence, not all viral vector-based vaccines will necessarily lead to protective responses for all immunogens and against all pathogens. This challenges the plug-and-play or one-size-fits-all aims of vaccine platforms for pandemic preparedness and underlines the need for a panel of alternative platforms. Two examples are AAV and VSV. AAV prior to vaccine engineering was poorly immunogenic due to inefficient uncoating in dendritic cells and so was more suitable for gene therapy [71]. VSV was only weakly immunogenic for the SARS-CoV-2 insert but is licensed as a vaccine against Ebola [62]. For HIV-1 prevention, vaccines stimulating de-novo responses may require a strong pro-inflammatory stimulation, but once the control of HIV-1 is established, therapeutic vaccines aiming to achieve post-ART HIV-1 remission may benefit from more ‘silent’ non-inflammatory vectors [72]. At the same time, strategies reactivating latent provirus reservoir must activate NF-κB, because HIV-1 transcription is strictly dependant on this transcription factor.

The best example of unique fine-tuned protection against a pathogenic SIV challenge is the consistent protection of approximately 55% of rhesus macaques achieved by particular molecular clone 68–1 of rhesus cytomegalovirus (rCMV) expressing SIV immunogens [73]. This vector carries five specific deletions in the viral genome, which completely prevent classical macaque major histocompatibility complex class Ia Mamu-A, B and C presentation of epitopes and instead induce nonclassical class Ib Mamu-E and class II-restricted CD8+ T cells. The mechanisms of this protection are being gradually deciphered [74▪▪,75,76], but it is species specific, as the same rhesus CMV vaccine does not protect cynomolgus macaques. Engineering of its human CMV equivalent has been attempted and candidate vaccine VIR-1111 is currently being evaluated in a phase 1 clinical trial in humans (NCT04725877).

CONCLUSION

Development of an effective vaccine against the most difficult of viruses, HIV-1, has been extremely challenging. The COVID-19 pandemic has momentarily focused the minds of politicians and funders on vaccinology and HIV-1 can benefit from this just like the SARS-CoV-2 vaccines benefited from decades of HIV-1 research. Novel vaccine vectors for subunit vaccines are an essential component of eventual success, which is now more in our grasp than it has ever been before.

Acknowledgements

The author would like to acknowledge the stimulating environment of the Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford.

Financial support and sponsorship

The author declares funding from the National Institutes of Health, Division of AIDS (Prime Contract No.: HHSN27220110002II/HHSN27200037, Subcontract No.: OX-14007–004–0037–212); the Medical Research Council U.K. and the U.K. Department for International Development under the MRC/DFID Concordat agreements (MR/N023668/1); the European and Developing Countries Clinical Trials Partnership (SRIA2015–1066); the European Union's Horizon 2020 Research and Innovation programme (grant agreement no. 681137-EAVI2020); and the AIDS International Collaborative Research Project of Joint Research Center for Human Retrovirus Infection, Kumamoto University supported by the Ministry of Education, Culture, Sports, Science and Technology of Japan.

Conflicts of interest

The author is one of the inventors on EU patent EP14846993.5 and U.S. patent PCT/US14/58422 concerning the HIVconsvX immunogen.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

▪ of special interest

▪▪ of outstanding interest

REFERENCES 1. Administration USFD. Vaccine licensed for use in the United States. 2022. https://www.fda.gov/vaccines-blood-biologics/vaccines/vaccines-licensed-use-united-states. [Accessed 23 July 2022]. 2. WHO. Novel COVID-19 vaccine tracker and landscape. 2022. https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines. [Accessed 23 July 2022]. 3. Baden LR, Stieh DJ, Sarnecki M, et al. Safety and immunogenicity of two heterologous HIV vaccine regimens in healthy, HIV-uninfected adults (TRAVERSE): a randomised, parallel-group, placebo-controlled, double-blind, phase 1/2a study. Lancet HIV 2020; 7:e688–e698. 4. Gray GE, Bekker LG, Laher F, et al. Vaccine efficacy of ALVAC-HIV and bivalent subtype C gp120-MF59 in adults. N Engl J Med 2021; 384:1089–1100. 5. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 2009; 361:2209–2220. 6. Zolla-Pazner S, Michael NL, Kim JH. A tale of four studies: HIV vaccine immunogenicity and efficacy in clinical trials. Lancet HIV 2021; 8:e449–e452. 7. Munson P. Progress towards a therapeutic HIV DNA vaccine. Expert Rev Vaccines 2022; 21:783–798. 8. Hanke T. Aiming for protective T-cell responses: a focus on the first generation conserved-region HIVconsv vaccines in preventive and therapeutic clinical trials. Expert Rev Vaccines 2019; 18:1029–1041. 9. Korber B, Fischer W. T cell-based strategies for HIV-1 vaccines. Hum Vaccin Immunother 2020; 16:713–722. 10. Hanke T. Conserved immunogens in prime-boost strategies for the next-generation HIV-1 vaccines. Expert Opin Biol Ther 2014; 14:601–616. 11▪. Aliahmad P, Miyake-Stoner SJ, Geall AJ, et al. Next generation self-replicating RNA vectors for vaccines and immunotherapies. Cancer Gene Ther 2022; [Online ahead of print]. 12. Mu Z, Haynes BF, Cain DW. Strategies for eliciting multiple lineages of broadly neutralizing antibodies to HIV by vaccination. Curr Opin Virol 2021; 51:172–178. 13. Reed SG, Bertholet S, Coler RN, et al. New horizons in adjuvants for vaccine development. Trends Immunol 2009; 30:23–32. 14. Fellay J, Shianna KV, Ge D, et al. A whole-genome association study of major determinants for host control of HIV-1. Science 2007; 317:944–947. 15. Pereyra F, Jia X, McLaren PJ, et al. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 2010; 330:1551–1557. 16. Rowland-Jones SL, McMichael A. Immune responses in HIV-exposed seronegatives: have they repelled the virus? Curr Opin Immunol 1995; 7:448–455. 17. McMichael AJ, Haynes BF. Lessons learned from HIV-1 vaccine trials: new priorities and directions. Nat Immunol 2012; 13:423–427. 18. Niessl J, Baxter AE, Mendoza P, et al. Combination anti-HIV-1 antibody therapy is associated with increased virus-specific T cell immunity. Nat Med 2020; 26:222–227. 19. Barbier AJ, Jiang AY, Zhang P, et al. The clinical progress of mRNA vaccines and immunotherapies. Nat Biotechnol 2022; 40:840–854. 20. Pardi N, Hogan MJ, Porter FW, et al. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov 2018; 17:261–279. 21. Weng Y, Li C, Yang T, et al. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv 2020; 40:107534. 22. van de Berg D, Kis Z, Behmer CF, et al. Quality by design modelling to support rapid RNA vaccine production against emerging infectious diseases. NPJ Vaccines 2021; 6:65. 23. Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med 2021; 384:403–416. 24. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med 2020; 383:2603–2615. 25. Sahin U, Kariko K, Tureci O. mRNA-based therapeutics: developing a new class of drugs. Nat Rev Drug Discov 2014; 13:759–780. 26. Hajj KA, Whitehead KA. Tools for translation: nonviral materials for therapeutic mRNA delivery. Nat Rev Mater 2017; 2:17056. 27. WHO. Messenger mRNA encoding the full-length SARS-CoV-2 spike glycoprotein. 2020. https://archive.org/web/20210105162941/https//mednet-communities.net/inn/db/media/docs/11889.doc. [Accessed 23 July 2022]. 28▪▪. Xia X. Detailed dissection and critical evaluation of the Pfizer/BioNTech and Moderna mRNA vaccines. Vaccines (Basel) 2021; 9:734. 29. Beaudoin CA, Bartas M, Volna A, et al. Are there hidden genes in DNA/RNA vaccines? Front Immunol 2022; 13:801915. 30. Anderson BR, Muramatsu H, Nallagatla SR, et al. Incorporation of pseudouridine into mRNA enhances translation by diminishing PKR activation. Nucleic Acids Res 2010; 38:5884–5892. 31. Kariko K, Buckstein M, Ni H, et al. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 2005; 23:165–175. 32. Gagnon P. Purification of nucleic acids. A handbook for purification of plasmid DNA and mRNA for gene therapy and vaccines. Vol. First electronic edition. Ajdovscina, Slovenia: BIA Separations; 2020. 33. Chen S, Ouyang H, He D, et al. Functionalized PAMAM-based nanoformulation for targeted delivery of 5-fluorouracil in hepatocellular carcinoma. Curr Pharm Des 2022; 28:2113–2125. 34. da Silva LT, Santillo BT, de Almeida A, et al. Using dendritic cell-based immunotherapy to treat HIV: how can this strategy be improved? Front Immunol 2018; 9:2993. 35. Mothe B, Hu X, Llano A, et al. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J Transl Med 2015; 13:60. 36. Van Lint S, Goyvaerts C, Maenhout S, et al. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res 2012; 72:1661–1671. 37. Leal L, Guardo AC, Moron-Lopez S, et al. Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection. AIDS 2018; 32:2533–2545. 38. Jong W, Leal L, Buyze J, et al. Therapeutic vaccine in chronically HIV-1-infected patients: a randomized, double-blind, placebo-controlled phase IIa trial with HTI-TriMix. Vaccines (Basel) 2019; 7:209. 39. Steichen JM, Kulp DW, Tokatlian T, et al. HIV vaccine design to target germline precursors of glycan-dependent broadly neutralizing antibodies. Immunity 2016; 45:483–496. 40. Ulmer JB, Deck RR, Dewitt CM, et al. Generation of MHC class I-restricted cytotoxic T lymphocytes by expression of a viral protein in muscle cells: antigen presentation by nonmuscle cells. Immunology 1996; 89:59–67. 41. Estcourt MJ, McMichael AJ, Hanke T. DNA vaccines against human immunodeficiency virus type 1. Immunol Rev 2004; 199:144–155. 42. Jacobson JM, Zheng L, Wilson CC, et al. The safety and immunogenicity of an interleukin-12-enhanced multiantigen DNA vaccine delivered by electroporation for the treatment of HIV-1 infection. J Acquir Immune Defic Syndr 2016; 71:163–171. 43. Rosenberg ES, Graham BS, Chan ES, et al. Safety and immunogenicity of therapeutic DNA vaccination in individuals treated with antiretroviral therapy during acute/early HIV-1 infection. PLoS One 2010; 5:e10555. 44. Roy MJ, Wu MS, Barr LJ, et al. Induction of antigen-specific CD8+ T cells, T helper cells, and protective levels of antibody in humans by particle-mediated administration of a hepatitis B virus DNA vaccine. Vaccine 2000; 19:764–778. 45. Mutua G, Farah B, Langat R, et al. Broad HIV-1 inhibition in vitro by vaccine-elicited CD8+ T cells in African adults. Mol Ther Methods Clin Dev 2016; 3:16061. 46. Bins AD, Jorritsma A, Wolkers MC, et al. A rapid and potent DNA vaccination strategy defined by in vivo monitoring of antigen expression. Nat Med 2005; 11:899–904. 47. Du X, Wang J, Zhou Q, et al. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv 2018; 25:1516–1525. 48. Ma Y, Tao W, Krebs SJ, et al. Vaccine delivery to the oral cavity using coated microneedles induces systemic and mucosal immunity. Pharm Res 2014; 31:2393–2403. 49. Rodriguez B, Asmuth DM, Matining RM, et al. Safety, tolerability, and immunogenicity of repeated doses of dermavir, a candidate therapeutic HIV vaccine, in HIV-infected patients receiving combination antiretroviral therapy: results of the ACTG 5176 trial. J Acquir Immune Defic Syndr 2013; 64:351–359. 50. Borthwick NJ, Lane T, Moyo N, et al. Randomized phase I trial HIV-CORE 003: Depletion of serum amyloid P component and immunogenicity of DNA vaccination against HIV-1. PLoS One 2018; 13:e0197299. 51. Xu Z, Patel A, Tursi NJ, et al. Harnessing recent advances in synthetic DNA and electroporation technologies for rapid vaccine development against COVID-19 and other emerging infectious diseases. Front Med Technol 2020; 2:571030. 52. De Rosa SC, Edupuganti S, Huang Y, et al. Robust antibody and cellular responses induced by DNA-only vaccination for HIV. JCI Insight 2020; 5:e137079. 53. Borthwick N, Ahmed T, Ondondo B, et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther 2014; 22:464–475. 54. Goonetilleke N, Moore S, Dally L, et al. Prime-boost vaccination with recombinant DNA and MVA expressing HIV-1 Clade A gag and immunodominant CTL epitopes induces multifunctional HIV-1-specific T cells in healthy subjects. J Virol 2006; 80:4717–4728. 55. Moyo N, Borthwick NJ, Wee EG, et al. Long-term follow up of human T-cell responses to conserved HIV-1 regions elicited by DNA/simian adenovirus/MVA vaccine regimens. PLoS One 2017; 12:e0181382. 56. Hammer SM, Sobieszczyk ME, Janes H, et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med 2013; 369:2083–2092. 57. Chen H, Xiang ZQ, Li Y, et al. Adenovirus-based vaccines: comparison of vectors from three families of adenovirideae. J Virol 2010; 84:10522–10532. 58. Gray GE, Moodie Z, Metch B, et al. Recombinant adenovirus type 5 HIV gag/pol/nef vaccine in South Africa: unblinded, long-term follow-up of the phase 2b HVTN 503/Phambili study. Lancet Infect Dis 2014; 14:388–396. 59. Ondondo B, Murakoshi H, Clutton G, et al. Novel conserved-region T-cell mosaic vaccine with high global HIV-1 coverage is recognized by protective responses in untreated infection. Mol Ther 2016; 24:832–842. 60. Agrati C, Capone S, Castilletti C, et al. Strong immunogenicity of heterologous prime-boost immunizations with the experimental vaccine GRAd-COV2 and BNT162b2 or ChAdOx1-nCOV19. NPJ Vaccines 2021; 6:131. 61. Lanini S, Capone S, Antinori A, et al. GRAd-COV2, a gorilla adenovirus-based candidate vaccine against COVID-19, is safe and immunogenic in younger and older adults. Sci Transl Med 2022; 14:eabj1996. 62. Suder E, Furuyama W, Feldmann H, et al. The vesicular stomatitis virus-based Ebola virus vaccine: from concept to clinical trials. Hum Vaccin Immunother 2018; 14:2107–2113. 63. Martinez-Navio JM, Fuchs SP, Pantry SN, et al. Adeno-associated virus delivery of anti-HIV monoclonal antibodies can drive long-term virologic suppression. Immunity 2019; 50:567–575. e5. 64. Blasi M, Wescott EC, Baker EJ, et al. Therapeutic vaccination with IDLV-SIV-Gag results in durable viremia control in chronically SHIV-infected macaques. NPJ Vaccines 2020; 5:36. 65. Wee EG, Ondondo B, Berglund P, et al. HIV-1 conserved mosaics delivered by regimens with integration-deficient DC-targeting lentiviral vector induce robust T cells. Mol Ther 2017; 25:494–503. 66. Travieso T, Li J, Mahesh S, et al. The use of viral vectors in vaccine development. NPJ Vaccines 2022; 7:75. 67. Folegatti PM, Ewer KJ, Aley PK, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020; 396:467–478. 68. Graham FL, van der Eb AJ. A new technique for the assay of infectivity of human adenovirus 5 DNA. Virology 1973; 52:456–467. 69▪▪. Almuqrin A, Davidson AD, Williamson MK, et al. SARS-CoV-2 vaccine ChAdOx1 nCoV-19 infection of human cell lines reveals low levels of viral backbone gene transcription alongside very high levels of SARS-CoV-2 S glycoprotein gene transcription. Genome Med 2021; 13:43. 70. Donovan-Banfield I, Turnell AS, Hiscox JA, et al. Deep splicing plasticity of the human adenovirus type 5 transcriptome drives virus evolution. Commun Biol 2020; 3:124. 71. Rossi A, Dupaty L, Aillot L, et al. Vector uncoating limits adeno-associated viral vector-mediated transduction of human dendritic cells and vector immunogenicity. Sci Rep 2019; 9:3631. 72. Goulder P, Deeks SG. HIV control: is getting there the same as staying there? PLoS Pathog 2018; 14:e1007222. 73. Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 2011; 473:523–527. 74▪▪. Barrenas F, Hansen SG, Law L, et al. Interleukin-15 response signature predicts RhCMV/SIV vaccine efficacy. PLoS Pathog 2021; 17:e1009278. 75. Hansen SG, Hancock MH, Malouli D, et al. Myeloid cell tropism enables MHC-E-restricted CD8(+) T cell priming and vaccine efficacy by the RhCMV/SIV vaccine. Sci Immunol 2022; 7:eabn9301. 76. Verweij MC, Hansen SG, Iyer R, et al. Modulation of MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine efficacy. Science 2021; 372:eabe9233.

留言 (0)

沒有登入
gif