npj Vaccines

A highly immunogenic UVC inactivated Sabin based polio vaccine
Tobin GJ, Tobin JK, Wiggins TJ, Bushnell RV, Kozar AV, Maale MF, MacLeod DA, Meeks HN, Daly MJ and Dollery SJ
Despite their efficacy, the currently available polio vaccines, oral polio vaccine (OPV) and inactivated polio vaccine (IPV), possess inherent flaws posing significant challenges in the global eradication of polio. OPV, which uses live Sabin attenuated strains, carries the risk of reversion to pathogenic forms and causing vaccine-associated paralytic poliomyelitis (VAPP) and vaccine-derived polio disease (VDPD) in incompletely vaccinated or immune-compromised individuals. Conventional IPVs, which are non-replicative, are more expensive to manufacture and introduce biohazard and biosecurity risks due to the use of neuropathogenic strains in production. These types of limitations have led to a call by the Global Polio Eradication Initiative and others for the development of updated polio vaccines. We are developing a novel Ultraviolet-C radiation (UVC) inactivation method that preserves immunogenicity and is compatible with attenuated strains of polio. The method incorporates an antioxidant complex, manganese-decapeptide-phosphate (MDP), derived from the radioresistant bacterium Deinococcus radiodurans. The inclusion of MDP protects the immunogenic neutralizing epitopes from damage during UVC inactivation. The novel vaccine candidate, ultraIPV, produced using these methods demonstrates three crucial attributes: complete inactivation, which precludes the risk of vaccine-associated disease; use of non-pathogenic strains to reduce production risks; and significantly enhanced yield of doses per milligram of input virus, which could increase vaccine supply while reducing costs. Additionally, ultraIPV retains antigenicity post-freeze-thaw cycles, a testament to its robustness.
A multi-epitope subunit vaccine providing broad cross-protection against diverse serotypes of Streptococcus suis
Liu J, Zhang Z, Pu W, Pan X, Li P, Bai Q, Liang S, Li C, Yu Y, Yao H and Ma J
Streptococcus suis infection represents a major challenge in pig farming and public health due to its zoonotic potential and diverse serotypes, while existing vaccines lack effective cross-protection. This study employed reverse vaccinology and immunoinformatics to identify 8 conserved proteins across 11 prevalent serotypes of S. suis. 16 candidate epitopes were selected to design three multi-epitope antigens against S. suis (designated as MEASs), which fused with a dendritic cell-targeting peptide to improve antigen presentation in host. Purified MEASs displayed favorable cross-reactogenicity against 29 serotype-specific antiserums. Robust humoral and cellular immune responses can be induced by MEAS 1 and MEAS 3 in a mouse model, which provided substantial protection against virulent strains from two different serotypes. In particular, their immune serums exhibited positive opsonization effects within bloodstream and macrophage phagocytosis. Taken together, we identified two promising MEASs with excellent cross-protection, offering potential in preventing S. suis infections in a mouse model.
Implications of the apparent extinction of B/Yamagata-lineage human influenza viruses
Barr IG and Subbarao K
Following the onset of the COVID-19 pandemic in 2020, the number of influenza viruses circulating globally fell to historically low numbers. Although influenza A and B/Victoria lineage viruses returned to normal patterns by 2022, B/Yamagata-lineage viruses have not been identified since 2020. The implications of the apparent extinction of this lineage of viruses on vaccine composition, and the risk of their re-introduction into the human population are discussed.
Mapping global public perspectives on mRNA vaccines and therapeutics
Xu J, Wu Z, Wass L, Larson HJ and Lin L
The development and rollout of mRNA vaccines during COVID-19 marked a significant advancement in vaccinology, yet public hesitation to vaccination was prevalent, indicating the potential risk that future mRNA-based medical innovations will fail to be adopted. Utilizing a combined approach of large language models with manual validation and unsupervised machine learning, we conducted a social listening analysis to assess attitudes towards mRNA vaccines and therapeutics on Twitter from June 2022 to May 2023, contrasting online perspectives with data from the Vaccine Adverse Event Reporting System. Our findings reveal widespread negative sentiment and a global lack of confidence in the safety, effectiveness, and trustworthiness of mRNA vaccines and therapeutics, with frequent discussions of severe vaccine side effects, rumors, and misinformation. This underscores the need for targeted communication strategies to foster acceptance of medical treatments and strengthen public trust in order to enhance societal resilience to future health challenges.
CRISPR/Cas9 screens identify key host factors that enhance rotavirus reverse genetics efficacy and vaccine production
Zhu Y, Sullender ME, Campbell DE, Wang L, Lee S, Kawagishi T, Hou G, Dizdarevic A, Jais PH, Baldridge MT and Ding S
Rotaviruses pose a significant threat to young children. To identify novel pro- and anti-rotavirus host factors, we performed genome-wide CRISPR/Cas9 screens using rhesus rotavirus and African green monkey cells. Genetic deletion of either SERPINB1 or TMEM236, the top two antiviral factors, in MA104 cells increased virus titers in a rotavirus strain independent manner. Using this information, we optimized the existing rotavirus reverse genetics systems by combining SERPINB1 knockout MA104 cells with a C3P3-G3 helper plasmid. We improved the recovery efficiency and rescued several low-titer rotavirus reporter and mutant strains that prove difficult to rescue otherwise. Furthermore, we demonstrate that TMEM236 knockout in Vero cells supported higher yields of two live-attenuated rotavirus vaccine strains than the parental cell line and represents a more robust vaccine-producing cell substrate. Collectively, we developed a third-generation optimized rotavirus reverse genetics system and generated gene-edited Vero cells as a new substrate for improving rotavirus vaccine production.
Preclinical characterization of the Omicron XBB.1.5-adapted BNT162b2 COVID-19 vaccine
Modjarrad K, Che Y, Chen W, Wu H, Cadima CI, Muik A, Maddur MS, Tompkins KR, Martinez LT, Cai H, Ramos M, Mensah S, Cumbia B, Falcao L, McKeen AP, Chang JS, Fennell KF, Huynh KW, McLellan TJ, Sahasrabudhe PV, Chen W, Cerswell M, Garcia MA, Li S, Sharma R, Li W, Dizon KP, Duarte S, Gillett F, Smith R, Illenberger DM, Efferen KS, Vogel AB, Anderson AS, Şahin U and Swanson KA
As SARS-CoV-2 evolves, increasing in potential for greater transmissibility and immune escape, updated vaccines are needed to boost adaptive immunity to protect against COVID-19 caused by circulating strains. Here, we report features of the monovalent Omicron XBB.1.5-adapted BNT162b2 vaccine, which contains XBB.1.5-specific sequence changes, relative to the original BNT162b2 backbone, in the encoded prefusion-stabilized SARS-CoV-2 spike protein (S(P2)). Biophysical characterization of Omicron XBB.1.5 S(P2) demonstrated that it maintains a prefusion conformation and adopts a flexible, predominantly open, state, with high affinity for the human ACE-2 receptor. When administered as a 4th dose in BNT162b2-experienced mice, the monovalent Omicron XBB.1.5 vaccine elicited substantially higher serum neutralizing titers against pseudotyped viruses of Omicron XBB.1.5, XBB.1.16, XBB.1.16.1, XBB.2.3, EG.5.1 and HV.1 sublineages and phylogenetically distant BA.2.86 lineage than the bivalent Wild Type + Omicron BA.4/5 vaccine. Similar trends were observed against Omicron XBB sublineage pseudoviruses when the vaccine was administered as a 2-dose series in naive mice. Strong S-specific Th1 CD4 and IFNγ CD8 T cell responses were also observed. These findings, together with real world performance of the XBB.1.5-adapted vaccine, suggest that preclinical data for the monovalent Omicron XBB.1.5-adapted BNT162b2 was predictive of protective immunity against dominant SARS-CoV-2 strains.
Obesity does not influence SARS-CoV-2 humoral vaccine immunogenicity
D'Souza M, Keeshan A, Gravel CA, Langlois MA and Cooper CL
Obesity is a recognized factor influencing immune function and infectious disease outcomes. Characterization of the influence of obesity on SARS-CoV-2 humoral vaccine immunogenicity is required to properly tailor vaccine type (mRNA, viral-vector, protein subunit vaccines) and dosing schedule. Data from a prospective cohort study collected over 34 months was used to evaluate the slope of antibody production and decay and neutralizing capacity following SARS-CoV-2 vaccination in individuals with and without obesity at baseline. Most participants were female (65.4%), white (92.4%), and received mRNA vaccines. 210 were obese and 697 non-obese. Sex and infection-acquired immunity were identified as effect modifiers for the relationship between obesity and COVID-19 vaccine humoral immunogenicity. No consistent influence of obesity on peak titres, titre retention, antibody isotype (IgG, IgM, IgA), or neutralization was identified when controlling for other key variables. It may not be necessary to consider this variable when developing SARS-CoV-2 vaccine dosing strategies.
Neutralization of SARS-CoV-2 KP.1, KP.1.1, KP.2 and KP.3 by human and murine sera
Xu K, An Y, Liu X, Xie H, Li D, Yang T, Duan M, Wang Y, Zhao X, Dai L and Gao GF
We report SARS-CoV-2 KP.1, KP.1.1, KP.2 and KP.3 neutralizing antibody titers. They displayed increased immune evasion compared to JN.1, especially KP.1 and KP.3, for participants who experienced BF.7/BA.5.2 breakthrough infection or received bivalent (delta/BA.5) vaccine boosting. Second XBB sub-variants breakthrough infection enhanced the neutralization responses. HK.3-JN.1 RBD-heterodimer induced balanced and potent neutralizing responses against recently-circulating SARS-CoV-2 sub-variants in mice, supporting to replace the COVID-19 antigen containing JN.1 or its sub-variants.
Probing novel epitopes on the Plasmodium falciparum circumsporozoite protein for vaccine development
Krenger PS, Roques M, Vogt AS, Pardini A, Rothen DA, Balke I, Schnider ST, Mohsen MO, Heussler VT, Zeltins A and Bachmann MF
RTS,S and R21 are the only vaccines recommended by the WHO to protect children from Plasmodium falciparum (Pf) clinical malaria. Both vaccines target the Pf sporozoite surface protein circumsporozoite protein (CSP). Recent studies showed that human antibodies neutralize Pf sporozoites most efficiently when simultaneously binding to the PfCSP NANP repeat and the NPDP junction domain. However, neither RTS,S nor R21 targets this junction domain. To test the potential of the NPDP junction domain and other sites of PfCSP as innovative vaccine targets, we developed multiple vaccine candidates based on cucumber mosaic virus-like particles (CuMV-VLPs). These candidates vary in several aspects: the number of targeted NANP repeats, the presence or absence of the junction domain, the cleavage site, and up to three NVDP repeats within the target sequence. Immunogenicity and efficacy studies were conducted in BALB/c mice, utilizing chimeric Plasmodium berghei (Pb) sporozoites, in which the endogenous CSP has been replaced by PfCSP (Pb/PfCSP). We observed a positive association between the number of targeted NANP repeats and the induction of specific IgM/IgG antibodies. Elevated humoral responses led to enhanced protection against parasitemia after Pb/PfCSP sporozoite challenge. Especially high-avidity/affinity antibody formation and vaccine protection were NANP repeat-dependent. Intriguingly, vaccine efficacy was not enhanced by targeting sites on PfCSP other than the NANP repeats. Our data emphasize the dominant role of the NANP repeat region for induction of protective antibodies. Furthermore, we present here novel malaria vaccine candidates with an excellent immunogenic profile that confer sterile protection in mice, even in absence of adjuvants.
NHBA antibodies elicited by 4CMenB vaccination are key for serum bactericidal activity against Neisseria gonorrhoeae
Tzeng YL, Sannigrahi S and Stephens DS
The 4CMenB (Bexsero) vaccine contains detergent-extracted outer membrane vesicles (OMVs) from a Neisseria meningitidis (Nm) group B strain NZ98/254 and three recombinant Nm protein antigens: Neisseria adhesin A (NadA), Factor H binding protein (FHbp, as the C-terminal protein in the GNA2091-FHbp fusion), and Neisserial Heparin Binding Antigen (NHBA, as the N-terminal protein in the NHBA-GNA1030 fusion). Previous work has shown that 4CMenB generates serum antibodies to Nm and Neisseria gonorrhoeae (Ng) OMV proteins and lipooligosaccharide (LOS). Mounting evidence indicates 4CMenB can partially protect against mucosal infections with Ng. The immunologic basis for Ng cross protection remains to be fully elucidated. Ten paired human sera obtained pre- and post-immunization with 4CMenB (1 month after a third vaccine dose) were used in ELISAs and in Western blots to determine IgG and IgA serum responses to OMVs from Nm strain NZ98/254 (OMV) and two Ng strains, 1291 and CNG20 (OMV), and gonococcal recombinant NHBA (rNHBA) proteins. Post 4CMenB sera, but not pre-sera, showed strong IgG and variable IgA responses to the OMV but lower (2-11-fold difference in signal intensity) recognition of OMV. All post (not pre) 4CMenB sera showed strong IgG, but variable IgA, recognition of rNHBA by ELISAs and Western blots. Three post 4CMenB sera at 10% (v/v) concentration had serum bactericidal activity (SBA) against Ng strains 1291 and CNG20 (~30-40% killing), not seen in paired pre-sera. These data confirmed 4CMenB-induced cross-reactive functional antibody responses to Ng. In competitive SBA assays, in which sera were pre-incubated with rNHBA, minimal SBA against Nm strain NZ98/254 was titrated away. However, most of the SBA against Ng strains 1291 and CNG20 required NHBA-specific antibodies, and the Δnhba mutants were resistant to killing by post 4CMenB sera. Removing NHBA-specific and LOS-specific OMV antibodies simultaneously decreased SBA significantly more than the sum of removing individual antibodies alone, suggesting synergy between anti-NHBA and anti-OMV antibodies. Anti- NHBA antibodies induced by 4CMenB vaccination cross react with NHBA and substantially contribute to the bactericidal response toward Ng induced by the vaccine.
Author Correction: Potent and long-lasting humoral and cellular immunity against varicella zoster virus induced by mRNA-LNP vaccine
Bhattacharya A, Jan L, Burlak O, Li J, Upadhyay G, Williams K, Dong J, Rohrer H, Pynn M, Simon A, Kuhlmann N, Pustylnikov S, Melo MB and Dey AK
Impact of rotavirus vaccination in Malawi from 2012 to 2022 compared to model predictions
Pitzer VE, Ndeketa L, Asare EO, Hungerford D, Lopman BA, Jere KC and Cunliffe NA
Rotarix® vaccine was introduced into the Malawi national immunization program in October 2012. We analyzed data on children <5 years old hospitalized with acute gastroenteritis from January 2012 to June 2022, and compared to pre-vaccination data from 1997 to 2009. We estimated vaccine coverage before, during, and after the COVID-19 pandemic using data from rotavirus-negative children. We compared the observed weekly number of rotavirus-associated gastroenteritis (RVGE) cases by age to predictions from a previously developed mathematical model to estimate overall vaccine effectiveness. The number of RVGE and rotavirus-negative acute gastroenteritis cases declined substantially following vaccine introduction. Vaccine coverage among rotavirus-negative controls was >90% with two doses by July 2014, and declined to a low of ~80% in October 2020 before returning to pre-pandemic levels by July 2021. Our models captured the post-vaccination trends in RVGE incidence. Comparing observed RVGE cases to the model-predicted incidence without vaccination, overall effectiveness was estimated to be modest at 36.0% (95% prediction interval: 33.6%, 39.9%), peaking in 2014, and was highest in infants (52.5%; 95% prediction interval: 50.1%, 54.9%). Our mathematical models provide a validated platform for assessing strategies to improve rotavirus vaccine impact in low-income settings.
Boosting neuraminidase immunity in the presence of hemagglutinin with the next generation of influenza vaccines
Cortés G, Ustyugova I, Farrell T, McDaniel C, Britain C, Romano C, N'Diaye S, Zheng L, Ferdous M, Iampietro J, Pougatcheva S, La Rue L, Han L, Ma F, Stegalkina S, Ray S, Zhang J and Barro M
Neuraminidase (NA), the second most abundant surface glycoprotein on the influenza virus, plays a key role in viral replication and propagation. Despite growing evidence showing that NA-specific antibodies correlate with resistance to disease in humans, current licensed vaccines focus almost entirely on the hemagglutinin (HA) antigen. Here, we demonstrate that recombinant NA (rNA) protein is highly immunogenic in both naïve mice and ferrets, as well as in pre-immune ferrets, irrespective of the level of match with preexisting immunity. Ferrets vaccinated with rNA developed mild influenza disease symptoms upon challenge with human H3N2 influenza virus, and anti-NA antibody responses appeared correlated with reduction in disease severity. The addition of rNA to a quadrivalent HA-based vaccine induced robust NA-specific humoral immunity in ferrets, while retaining the ability to induce HA-specific immunity. These results demonstrate that the addition of rNA is a viable option to increase immunogenicity and potentially efficacy versus currently licensed influenza vaccines by means of boosting NA immunity.
Irradiated whole cell Chlamydia vaccine confers significant protection in a murine genital tract challenge model
Broder KC, Matrosova VY, Tkavc R, Gaidamakova EK, Ho LTVT, Macintyre AN, Soc A, Diallo A, Darnell SC, Bash S, Daly MJ, Jerse AE and Liechti GW
Chlamydia trachomatis infections are the most common bacterial STIs globally and can lead to serious morbidity if untreated. Development of a killed, whole-cell vaccine has been stymied by coincident epitope destruction during inactivation. Here, we present a prototype Chlamydia vaccine composed of elementary bodies (EBs) from the related mouse pathogen, Chlamydia muridarum (Cm). EBs inactivated by gamma rays (Ir-Cm) in the presence of the antioxidant Mn-Decapeptide (DEHGTAVMLK) Phosphate (MDP) are protected from epitope damage but not DNA damage. Cm EBs gamma-inactivated with MDP retain their structure and provide significant protection in a murine genital tract infection model. Mice vaccinated with Ir-Cm (+MDP) exhibited elevated levels of Cm-specific IgG and IgA antibodies, reduced bacterial burdens, accelerated clearance, and distinctive cytokine responses compared to unvaccinated controls and animals vaccinated with EBs irradiated without MDP. Preserving EB epitopes with MDP during gamma inactivation offers the potential for a polyvalent, whole-cell vaccine against C. trachomatis.
A Lassa virus live attenuated vaccine candidate that is safe and efficacious in guinea pigs
Carey BD, Yu S, Geiger J, Ye C, Huzella LM, Reeder RJ, Mehta M, Hirsch S, Bernbaum R, Cubitt B, Pahar B, Anthony SM, Marketon A, Bernbaum JG, Tran JP, Crozier I, Martínez-Sobrido L, Worwa G, de la Torre JC and Kuhn JH
Lassa virus (LASV) is a rodent-borne mammarenavirus that causes tens to hundreds of thousands of human infections annually in Western Africa. Approximately 20% of these infections progress to Lassa fever (LF), an acute disease with case-fatality rates from ≈20-70%. Currently, there are no approved vaccines or specific therapeutics to prevent or treat LF. The LASV genome consists of a small (S) segment that has two genes, GP and NP, and a large (L) segment that has two genes, L and Z. In both segments, the two genes are separated by non-coding intergenic regions (IGRs). Recombinant LASVs (rLASVs), in which the L segment IGR was replaced with the S segment IGR or in which the GP gene was codon-deoptimized, lost fitness in vitro, were highly attenuated in vivo, and, when used as vaccines, protected domesticated guinea pigs from otherwise lethal LASV exposure. Here, we report the generation of rLASV/IGR-CD, which includes both determinants of attenuation and further enhances the safety of the vaccine compared with its predecessors. rLASV/IGR-CD grew to high titers in Vero cells, which are approved for human vaccine production, but did not cause signs of disease or pathology in guinea pigs. Importantly, guinea pigs vaccinated with rLASV/IGR-CD were completely protected from disease and death after a typically lethal exposure to wild-type LASV. Our data support the development of rLASV/IGR-CD as a live-attenuated LF vaccine with stringent safety features.
Improving precision of vaccine efficacy evaluation using immune correlate data in time-to-event models
Dudášová J, Valenta Z and Sachs JR
Understanding potential differences in vaccine-induced protection between demographic subgroups is key for vaccine development. Vaccine efficacy evaluation across these subgroups in phase 2b or 3 clinical trials presents challenges due to lack of precision: such trials are typically designed to demonstrate overall efficacy rather than to differentiate its value between subgroups. This study proposes a method for estimating vaccine efficacy using immunogenicity (instead of vaccination status) as a predictor in time-to-event models. The method is applied to two datasets from immunogenicity sub-studies of vaccine phase 3 clinical trials for zoster and dengue vaccines. Results show that using immunogenicity-based estimation of efficacy in subgroups using time-to-event models is more precise than the standard estimation. Incorporating immune correlate data in time-to-event models improves precision in estimating efficacy (i.e., yields narrower confidence intervals), which can assist vaccine developers and public health authorities in making informed decisions.
Preclinical evaluation of a universal inactivated influenza B vaccine based on the mosaic hemagglutinin-approach
González-Domínguez I, Puente-Massaguer E, Abdeljawad A, Lai TY, Liu Y, Loganathan M, Francis B, Lemus N, Dolange V, Boza M, Slamanig S, Martínez-Guevara JL, Krammer F, Palese P and Sun W
We have developed a new universal influenza B vaccination strategy based on inactivated influenza B viruses displaying mosaic hemagglutinins (mHAs). Recombinant mHA viruses were constructed by replacing the four major antigenic sites of influenza B virus HAs, with those from exotic avian influenza A virus HAs. Sequential vaccination of naïve mice with mHA-based vaccines elicited higher immune responses towards the immuno-subdominant conserved epitopes of the HA than vaccination with wildtype viruses. Among the different preparations tested, mHA split vaccines were less immunogenic than their whole inactivated virus counterparts. This lower immunogenicity was overcome by the combination with adjuvants. mHA split vaccines adjuvanted with a Toll-like receptor-9 agonist (CpG 1018) increased Th1 immunity and in vivo cross-protection, whereas adjuvanting with an MF59-like oil-in-water nano-emulsion (AddaVax) enhanced and broadened humoral immune responses and antibody-mediated cross-protection. The mHA vaccines with or without adjuvant were subsequently evaluated in mice that were previously immunized to closely mimic human pre-existing immunity to influenza B viruses and the contribution of innate and cellular immunity was evaluated in this model. We believe these preclinical studies using the mHA strategy represent a major step toward the evaluation of a universal influenza B virus vaccine in clinical trials.
Assessing acute kidney injury risk after COVID vaccination and infection in a large cohort study
Pan Y, Han Y, Zhou C, Zheng J, Zhao L, Ye X, He Y and
Acute kidney injury (AKI) has been noticed after both COVID-19 vaccination and infection, affecting risk-benefit evaluations and vaccine hesitancy. We conducted a large-scale N3C cohort study to compare AKI incidence following COVID-19 vaccination and infection. Participants from December 2020 to August 2023 were divided into two groups based on their initially observed COVID-19 antigen exposure: COVID-19 vaccination group (n = 2,953,219) and COVID-19 infection group (n = 3,616,802). AKI was defined by diagnostic codes and serum creatinine changes within a 30 day follow-up window after exposure. The absolute risk of AKI was 0.66% in the vaccination group versus 4.88% in the infection group. After adjusting for various confounders, COVID-19 infection was associated with a significantly higher risk of AKI than COVID-19 vaccination (aHR = 10.31, P < 0.001). Our study reveals that COVID-19 vaccination is associated with a significant lower AKI risk compared to COVID-19 infection.
Phase 1 clinical trial of Hantaan and Puumala virus DNA vaccines delivered by needle-free injection
Hooper JW, Kwilas SA, Josleyn M, Norris S, Hutter JN, Hamer M, Livezey J, Paolino K, Twomey P, Koren M, Keiser P, Moon JE, Nwaeze U, Koontz J, Ledesma-Feliciano C, Landry N and Wellington T
Hantaan virus (HTNV) and Puumala virus (PUUV) are pathogenic zoonoses found in Asia and Europe, respectively. We conducted a randomized Phase 1 clinical trial of individual HTNV and PUUV DNA vaccines targeting the envelope glycoproteins (GnGc), as well as a combined HTNV/PUUV DNA vaccine delivered at varying doses using the PharmaJet Stratis® needle-free injection system (NCT02776761). Cohort 1 and 2 vaccines consisted of 2 mg/vaccination of HTNV or PUUV plasmid, respectively. Cohort 3 vaccine consisted of 2 mg/vaccination of 1:1 mixture of HTNV and PUUV vaccines. Vaccinations were administered on Days 0, 28, 56, and 168. The vaccines were safe and well tolerated. Neutralizing antibody responses were elicited in 7/7 (100%) subjects who received the HTNV DNA (Cohort 1) and 6/6 (100%) subjects who received the PUUV DNA (Cohort 2) vaccines alone. The combination vaccine resulted in 4/9 (44%) seroconversion against both viruses. After the first two vaccinations, the seroconversion rates for the HTNV and PUUV vaccines were >80%.
Leishmania tarentolae as a platform for the production of vaccines against viral pathogens
Zimna M and Krol E
Infectious diseases remain a persistent public health problem and a leading cause of morbidity and mortality in both humans and animals. The most effective method of combating viral infections is the widespread use of prophylactic vaccinations, which are administered to both people at risk of disease and animals that may serve as significant sources of infection. Therefore, it is crucial to develop technologies for the production of vaccines that are highly effective, easy to transport and store, and cost-effective. The protein expression system based on the protozoan Leishmania tarentolae offers several advantages, validated by numerous studies, making it a good platform for producing vaccine antigens. This review provides a comprehensive overview into the potential applications of L. tarentolae for the safe production of effective viral antigens.
MTBVAC induces superior antibody titers and IgG avidity compared to BCG vaccination in non-human primates
Peralta-Álvarez MP, Downward K, White A, Redondo Azema H, Sibley L, Sarfas C, Morrison A, Dennis M, Diaz-Santana D, Harris SA, Li S, Puentes E, Aguilo N, Martin C, Sharpe S, McShane H and Tanner R
The only currently licensed vaccine against tuberculosis (TB), Bacille Calmette Guérin (BCG), is insufficient to control the epidemic. MTBVAC is a live attenuated strain of Mycobacterium tuberculosis (M.tb) and is one the most advanced TB vaccine candidates in the pipeline. It is more efficacious than BCG in preclinical models including non-human primates (NHPs), and has demonstrated safety and immunogenicity in human populations. To better understand the immune mechanisms underlying the superior efficacy conferred by MTBVAC, we characterized M.tb-specific antibody responses in NHPs vaccinated with either BCG or MTBVAC. MTBVAC vaccination induced higher titers of IgG, IgM and IgA, and higher avidity IgG compared with BCG vaccination. IgG avidity correlated with protection following M.tb challenge in the same animals, validating the association previously reported between this measure and protection in the context of intravenous BCG vaccination, suggesting that IgG avidity may represent a relevant marker or correlate of protection from TB.