Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (102)

Search Parameters:
Keywords = heterologous prime and boost

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 2688 KiB  
Article
Recombinant Tetrameric Neuraminidase Subunit Vaccine Provides Protection Against Swine Influenza A Virus Infection in Pigs
by Ao Zhang, Bin Tan, Jiahui Wang and Shuqin Zhang
Vaccines 2025, 13(8), 783; https://doi.org/10.3390/vaccines13080783 - 23 Jul 2025
Viewed by 355
Abstract
Background/Objectives: Swine influenza A virus (swIAV), a prevalent respiratory pathogen in porcine populations, poses substantial economic losses to global livestock industries and represents a potential threat to public health security. Neuraminidase (NA) has been proposed as an important component for universal influenza [...] Read more.
Background/Objectives: Swine influenza A virus (swIAV), a prevalent respiratory pathogen in porcine populations, poses substantial economic losses to global livestock industries and represents a potential threat to public health security. Neuraminidase (NA) has been proposed as an important component for universal influenza vaccine development. NA has potential advantages as a vaccine antigen in providing cross-protection, with specific antibodies that have a broad binding capacity for heterologous viruses. In this study, we evaluated the immunogenicity and protective efficacy of a tetrameric recombinant NA subunit vaccine in a swine model. Methods: We constructed and expressed structurally stable soluble tetrameric recombinant NA (rNA) and prepared subunit vaccines by mixing with ISA 201 VG adjuvant. The protective efficacy of rNA-ISA 201 VG was compared to that of a commercial whole inactivated virus vaccine. Pigs received a prime-boost immunization (14-day interval) followed by homologous viral challenge 14 days post-boost. Results: Both rNA-ISA 201 VG and commercial vaccine stimulated robust humoral responses. Notably, the commercial vaccine group exhibited high viral-binding antibody titers but very weak NA-specific antibodies, whereas rNA-ISA 201 VG immunization elicited high NA-specific antibody titers alongside substantial viral-binding antibodies. Post-challenge, both immunization with rNA-ISA 201 VG and the commercial vaccine were effective in inhibiting viral replication, reducing viral load in porcine respiratory tissues, and effectively mitigating virus-induced histopathological damage, as compared to the PBS negative control. Conclusions: These findings found that the anti-NA immune response generated by rNA-ISA 201 VG vaccination provided protection comparable to that of a commercial inactivated vaccine that primarily induces an anti-HA response. Given that the data are derived from one pig per group, there is a requisite to increase the sample size for more in-depth validation. This work establishes a novel strategy for developing next-generation SIV subunit vaccines leveraging NA as a key immunogen. Full article
(This article belongs to the Special Issue Vaccine Development for Swine Viral Pathogens)
Show Figures

Figure 1

25 pages, 4161 KiB  
Article
Systemic and Mucosal Antibody Responses to SARS-CoV-2 Variant-Specific Prime-and-Boost and Prime-and-Spike Vaccination: A Comparison of Intramuscular and Intranasal Bivalent Vaccine Administration in a Murine Model
by Mariam Maltseva, Yannick Galipeau, Pauline McCluskie, Nicolas Castonguay, Curtis L. Cooper and Marc-André Langlois
Vaccines 2025, 13(4), 351; https://doi.org/10.3390/vaccines13040351 - 25 Mar 2025
Viewed by 1179
Abstract
Background: The rapid genetic evolution of SARS-CoV-2 has led to the emergence of immune-evading, highly transmissible variants of concern (VOCs). This prompts the need for next-generation vaccines that elicit robust mucosal immunity in the airways to directly curb viral infection. Objective: Here, we [...] Read more.
Background: The rapid genetic evolution of SARS-CoV-2 has led to the emergence of immune-evading, highly transmissible variants of concern (VOCs). This prompts the need for next-generation vaccines that elicit robust mucosal immunity in the airways to directly curb viral infection. Objective: Here, we investigate the impact of heterologous variant prime–boost regimens on humoral responses, focusing on intramuscular (IM) and intranasal (IN) routes of administration. Using a murine model, we assessed the immunogenicity of unadjuvanted protein boosts with Wu-1, Omicron BA.4/5, or Wu-1 + BA.4/5 spike antigens following monovalent or bivalent IM priming with mRNA-LNP vaccines. Results: IM priming induced strong systemic total and neutralizing antibody responses that were further enhanced by IN boosts with BA.4/5. IN boosting achieved the broadest serum neutralization across all VOCs tested. Notably, bivalent mRNA-LNP IM priming induced robust, cross-variant serum neutralizing antibody production, independent of subsequent IN boost combinations. Conclusions: Our findings highlight the benefit of including distinct antigenic variants in the prime vaccination followed by a variant-tailored IN boost to elicit both systemic and mucosal variant-specific responses that are potentially capable of reducing SARS-CoV-2 transmission. Full article
Show Figures

Graphical abstract

17 pages, 3719 KiB  
Article
Decade-Long Sustained Cellular Immunity Induced by Sequential and Repeated Vaccination with Four Heterologous HIV Vaccines in Rhesus Macaques
by Xiaozhou He, Danying Chen, Qi Ma, Yanzhe Hao, Hongxia Li, Xiaoguang Zhang, Yuxi Cao and Xia Feng
Vaccines 2025, 13(4), 338; https://doi.org/10.3390/vaccines13040338 - 21 Mar 2025
Viewed by 712
Abstract
Background/Objectives: Developing durable cellular immunity remains a critical challenge for HIV vaccine development. Methods: We evaluated a sequential and repeated heterologous prime–boost vaccination regimen using four distinct vector-based vaccines (DNA, rAd5, rSeV, and rMVA) expressing HIV-1 gag in rhesus macaques over a decade-long [...] Read more.
Background/Objectives: Developing durable cellular immunity remains a critical challenge for HIV vaccine development. Methods: We evaluated a sequential and repeated heterologous prime–boost vaccination regimen using four distinct vector-based vaccines (DNA, rAd5, rSeV, and rMVA) expressing HIV-1 gag in rhesus macaques over a decade-long observation period. Results: Compared to the two-vector and control groups, the four-vector regimen elicited potent gag-specific cellular immune responses, as evidenced by IFN-γ ELISPOT assays showing sustained responses exceeding 500 SFCs/106 PBMCs for up to 52 or 69 weeks post-vaccination. Intracellular cytokine staining revealed multifunctional CD4+ and CD8+ T-cell responses, while humoral immunity against Ad5 vectors remained manageable despite repeated administrations. Conclusions: These findings demonstrate that sequential and repeated heterologous vaccination effectively induces and maintains durable cellular immunity, providing a strategic framework for HIV vaccine design. Full article
(This article belongs to the Section Vaccine Advancement, Efficacy and Safety)
Show Figures

Figure 1

13 pages, 710 KiB  
Article
Pooled Analysis of the Effect of Pre-Existing Ad5 Neutralizing Antibodies on the Immunogenicity of Adenovirus Type 5 Vector-Based COVID-19 Vaccine from Eight Clinical Trials
by Wenqing Liu, Yuqing Li, Xiaolong Li, Feiyu Wang, Runjie Qi, Tao Zhu and Jingxin Li
Vaccines 2025, 13(3), 333; https://doi.org/10.3390/vaccines13030333 - 20 Mar 2025
Viewed by 816
Abstract
Background: Pre-existing adenovirus immunity restricts the utilization of adenovirus-vectored vaccines. The current study aims to conduct a pooled analysis of eight clinical trials to evaluate the influence of pre-existing Ad5 neutralizing antibodies on immunogenicity of Ad5-nCoV. Methods: The primary outcome indicator [...] Read more.
Background: Pre-existing adenovirus immunity restricts the utilization of adenovirus-vectored vaccines. The current study aims to conduct a pooled analysis of eight clinical trials to evaluate the influence of pre-existing Ad5 neutralizing antibodies on immunogenicity of Ad5-nCoV. Methods: The primary outcome indicator of this pooled analysis is the geometric mean titers (GMTs) of live SARS-CoV-2 NAbs against the wild-type strain on day 28 post-vaccination. Participants were divided into two cohorts: an adolescent cohort comprising individuals aged 6–17 years and an adult cohort with individuals aged 18 years and older. Within each cohort, individuals were further categorized into three subgroups based on their Ad5-nCoV vaccination schedules: one subgroup received a single intramuscular dose as the primary regimen (Ad5-IM-prime), another received an intramuscular dose as the heterologous prime-boost regimen (Ad5-IM-boost), and the last subgroup received an aerosolized dose as the heterologous prime-boost regimen (Ad5-IH-boost). Results: A total of 3512 participants were included in this pooled analysis. In the Ad5-IM-prime subgroup, there were 1001 adolescents and 1450 adults; in the Ad5-IM-boost subgroup, there were 65 adolescents and 396 adults; and in the Ad5-IH-boost subgroup, there were 207 adolescents and 393 adults. In the adult cohort, the GMTs of NAbs against wild-type SARS-CoV-2 on day 28 post-vaccination for the Ad5-IM-prime, Ad5-IM-boost, and Ad5-IH-boost subgroups were 35.6 (95% CI: 32.0, 39.7), 358.3 (95% CI: 267.6, 479.6), and 2414.1 (95% CI: 2006.9, 2904.0), respectively, with negative (less than 1:12) pre-existing NAb titers compared to 10.7 (95% CI: 9.1, 12.6), 116.9 (95% CI: 84.9, 161.1), and 762.7 (95% CI: 596.2, 975.8), respectively, with high (greater than 1:1000) pre-existing NAb titers. A similar trend was observed in the adolescent cohort, where pre-existing immunity was found to reduce the peak of live SARS-CoV-2 Nabs post-vaccination. Conclusions: Regardless of whether Ad5-nCoV is administered as a primary vaccination regimen or as a heterologous prime-boost strategy, a negative impact on immunogenicity can still be observed in the presence of high pre-existing immunity. However, when primary immunization is achieved with inactivated COVID-19 vaccines, aerosol inhalation can significantly enhance the immunogenicity of Ad5-nCoV compared to intramuscular injections of Ad5-nCoV as a booster. Full article
Show Figures

Figure 1

19 pages, 2598 KiB  
Article
Prime-Boost Vaccination Based on Nanospheres and MVA Encoding the Nucleoprotein of Crimean-Congo Hemorrhagic Fever Virus Elicits Broad Immune Responses
by Eva Calvo-Pinilla, Sandra Moreno, Natalia Barreiro-Piñeiro, Juana M. Sánchez-Puig, Rafael Blasco, José Martínez-Costas, Alejandro Brun and Gema Lorenzo
Vaccines 2025, 13(3), 291; https://doi.org/10.3390/vaccines13030291 - 10 Mar 2025
Cited by 1 | Viewed by 1291
Abstract
Background/Objectives: Crimean–Congo hemorrhagic fever virus (CCHFV) is an emerging, widely distributed zoonotic tick-borne pathogen. The virus causes severe disease in humans, and numerous wild and domestic animals act as reservoirs of it. Unfortunately, there are no effective therapies or safe vaccines commercialized [...] Read more.
Background/Objectives: Crimean–Congo hemorrhagic fever virus (CCHFV) is an emerging, widely distributed zoonotic tick-borne pathogen. The virus causes severe disease in humans, and numerous wild and domestic animals act as reservoirs of it. Unfortunately, there are no effective therapies or safe vaccines commercialized nowadays for this particular virus. As CCHF (Crimean–Congo hemorrhagic fever) is a serious threat to public health, there is an urgent need to investigate the development of safe and effective vaccination strategies further. Methods: In this work, we have employed two immunization platforms based on protein nanoparticles and a modified vaccinia Ankara (MVA) viral vector using the nucleoprotein (NP) as the target antigen. The humoral and cellular immune responses were characterized by ELISA, ICS, and cytokine measurement. Results: This work shows that a single dose of the vaccine candidates was not as immunogenic as the heterologous vaccination using nanoparticles and MVA. A prime with NP nanoparticles (NS-NP) and a boost with MVA-expressing NP were capable of triggering significant levels of humoral and cellular immune responses against CCHFV in mice. Conclusions: Our study shows that the NS-NP/MVA-NP vaccination strategy effectively elicits a robust humoral and cellular immune response in a mouse model, emphasizing its potential as a protective approach against CCHFV lineages. Full article
(This article belongs to the Special Issue Veterinary Vaccines and Host Immune Responses)
Show Figures

Figure 1

23 pages, 4285 KiB  
Article
Computationally Selected Multivalent HIV-1 Subtype C Vaccine Protects Against Heterologous SHIV Challenge
by Dieter Mielke, Marina Tuyishime, Natasha S. Kelkar, Yunfei Wang, Robert Parks, Sampa Santra, Wes Rountree, LaTonya D. Williams, Tiffany Peters, Nathan Eisel, Sheetal Sawant, Lu Zhang, Derrick Goodman, Shalini Jha, Adam Zalaquett, Pratamesh Ramasubramanian, Sherry Stanfield-Oakley, Gary Matyas, Zoltan Beck, Mangala Rao, Julie Ake, Thomas N. Denny, David C. Montefiori, Margaret E. Ackerman, Lawrence Corey, Georgia D. Tomaras, Bette T. Korber, Barton F. Haynes, Xiaoying Shen and Guido Ferrariadd Show full author list remove Hide full author list
Vaccines 2025, 13(3), 231; https://doi.org/10.3390/vaccines13030231 - 24 Feb 2025
Cited by 1 | Viewed by 1036
Abstract
Background: The RV144 trial in Thailand is the only HIV-1 vaccine efficacy trial to date to demonstrate any efficacy. Genetic signatures suggested that antibodies targeting the variable loop 2 (V2) of the HIV-1 envelope played an important protective role. The ALVAC prime [...] Read more.
Background: The RV144 trial in Thailand is the only HIV-1 vaccine efficacy trial to date to demonstrate any efficacy. Genetic signatures suggested that antibodies targeting the variable loop 2 (V2) of the HIV-1 envelope played an important protective role. The ALVAC prime and protein boost follow-up trial in southern Africa (HVTN702) failed to show any efficacy. One hypothesis for this is the greater diversity of subtype C viruses in southern Africa relative to CRF01_AE in Thailand. Methods: Here, we determined whether an ALVAC prime with computationally selected gp120 boost immunogens maximizing coverage of diversity of subtype C viruses in the variable V1 and V2 regions (V1V2) improved the protection of non-human primates (NHPs) from a heterologous subtype C SHIV challenge compared to more traditional regimens. Results: An ALVAC prime with Trivalent subtype C gp120 boosts resulted in statistically significant protection from repeated intrarectal SHIV challenges compared to the control. Evaluation of the immunogenicity of each vaccine regimen at the time of challenge demonstrated that different gp120 combination boosts elicited similar high magnitudes of gp120 and breadth of V1V2-binding antibodies, as well as strong Fc-mediated immune responses. Low-to-no neutralization of the challenge virus was detected. A Cox proportional hazard analysis of five pre-selected immune parameters at the time of challenge identified ADCC against the challenge envelope as a correlate of protection. Systems serology analysis revealed that immune responses elicited by the different vaccine regimens were distinct and identified further correlates of resistance to infection. Conclusions: Computationally designed vaccines with maximized subtype C V1V2 coverage mediated protection of NHPs from a heterologous Tier-2 subtype C SHIV challenge. Full article
(This article belongs to the Special Issue Advances in HIV Vaccine Development)
Show Figures

Figure 1

19 pages, 8275 KiB  
Article
Adenoviral Vector-Based Vaccine Expressing Hemagglutinin Stem Region with Autophagy-Inducing Peptide Confers Cross-Protection Against Group 1 and 2 Influenza A Viruses
by Wen-Chien Wang, Ekramy E. Sayedahmed, Marwa Alhashimi, Ahmed Elkashif, Vivek Gairola, Muralimanohara S. T. Murala, Suryaprakash Sambhara and Suresh K. Mittal
Vaccines 2025, 13(1), 95; https://doi.org/10.3390/vaccines13010095 - 20 Jan 2025
Cited by 2 | Viewed by 1958
Abstract
Background/Objectives: An effective universal influenza vaccine is urgently needed to overcome the limitations of current seasonal influenza vaccines, which are ineffective against mismatched strains and unable to protect against pandemic influenza. Methods: In this study, bovine and human adenoviral vector-based vaccine platforms were [...] Read more.
Background/Objectives: An effective universal influenza vaccine is urgently needed to overcome the limitations of current seasonal influenza vaccines, which are ineffective against mismatched strains and unable to protect against pandemic influenza. Methods: In this study, bovine and human adenoviral vector-based vaccine platforms were utilized to express various combinations of antigens. These included the H5N1 hemagglutinin (HA) stem region or HA2, the extracellular domain of matrix protein 2 of influenza A virus, HA signal peptide (SP), trimerization domain, excretory peptide, and the autophagy-inducing peptide C5 (AIP-C5). The goal was to identify the optimal combination for enhanced immune responses and cross-protection. Mice were immunized using a prime-boost strategy with heterologous adenoviral (Ad) vectors. Results: The heterologous Ad vectors induced robust HA stem-specific humoral and cellular immune responses in the immunized mice. Among the tested combinations, Ad vectors expressing SP + HA stem + AIP-C5 conferred significant protection against group 1 (H1N1 and H5N1) and group 2 (H3N2) influenza A viruses. This protection was demonstrated by lower lung viral titers and reduced morbidity and mortality. Conclusions: The findings support further investigation of heterologous Ad vaccine platforms expressing SP + HA stem + AIP-C5. This combination shows promise as a potential universal influenza vaccine, providing broader protection against influenza A viruses. Full article
(This article belongs to the Special Issue Advances in Vaccines against Infectious Diseases)
Show Figures

Figure 1

19 pages, 3213 KiB  
Article
Intranasal Prime–Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection
by Muriel Metko, Jason Tonne, Alexa Veliz Rios, Jill Thompson, Haley Mudrick, David Masopust, Rosa Maria Diaz, Michael A. Barry and Richard G. Vile
Vaccines 2024, 12(10), 1191; https://doi.org/10.3390/vaccines12101191 - 18 Oct 2024
Viewed by 1930
Abstract
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime–boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon [...] Read more.
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime–boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. Methods: Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. Results: In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. Conclusions: There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Graphical abstract

23 pages, 3742 KiB  
Article
A Head-to-Head Comparative Study of the Replication-Competent Vaccinia Virus and AAV1-Based Malaria Vaccine versus RTS,S/AS01 in Murine Models
by Kartika Hardianti Zainal, Ammar Abdurrahman Hasyim, Yutaro Yamamoto, Tetsushi Mizuno, Yuna Sato, Sani Hadiyan Rasyid, Mamoru Niikura, Yu-ichi Abe, Mitsuhiro Iyori, Hiroaki Mizukami, Hisatoshi Shida and Shigeto Yoshida
Vaccines 2024, 12(10), 1155; https://doi.org/10.3390/vaccines12101155 - 10 Oct 2024
Cited by 1 | Viewed by 2126
Abstract
Background/Objectives: We developed a multistage Plasmodium falciparum vaccine using a heterologous prime-boost immunization strategy. This involved priming with a highly attenuated, replication-competent vaccinia virus strain LC16m8Δ (m8Δ) and boosting with adeno-associated virus type 1 (AAV1). This approach demonstrated 100% efficacy in both [...] Read more.
Background/Objectives: We developed a multistage Plasmodium falciparum vaccine using a heterologous prime-boost immunization strategy. This involved priming with a highly attenuated, replication-competent vaccinia virus strain LC16m8Δ (m8Δ) and boosting with adeno-associated virus type 1 (AAV1). This approach demonstrated 100% efficacy in both protection and transmission-blocking in a murine model. In this study, we compared our LC16m8∆/AAV1 vaccine, which harbors a gene encoding Pfs25-PfCSP fusion protein, to RTS,S/AS01 (RTS,S) in terms of immune responses, protective efficacy, and transmission-blocking activity (TBA) in murine models. Methods: Mice were immunized following prime-boost vaccine regimens m8∆/AAV1 or RTS,S and challenged with transgenic Plasmodium berghei parasites. Immune responses were assessed via ELISA, and TB efficacy was evaluated using direct feeding assays. Results: m8∆/AAV1 provided complete protection (100%) in BALB/c mice and moderate (40%) protection in C57BL/6 mice, similar to RTS,S. Unlike RTS,S’s narrow focus (repeat region), m8∆/AAV1 triggered antibodies for all PfCSP regions (N-terminus, repeat, and C-terminus) with balanced Th1/Th2 ratios. Regarding transmission blockade, serum from m8∆/AAV1-vaccinated BALB/c mice achieved substantial transmission-reducing activity (TRA = 83.02%) and TB activity (TBA = 38.98%)—attributes not observed with RTS,S. Furthermore, m8∆/AAV1 demonstrated durable TB efficacy (94.31% TRA and 63.79% TBA) 100 days post-immunization. Conclusions: These results highlight m8∆/AAV1′s dual action in preventing sporozoite invasion and onward transmission, a significant advantage over RTS,S. Consequently, m8∆/AAV1 represents an alternative and a promising vaccine candidate that can enhance malaria control and elimination strategies. Full article
(This article belongs to the Special Issue Parasitic Infections: Therapy for Host Immunity and Vaccination)
Show Figures

Figure 1

12 pages, 6798 KiB  
Article
Enhanced Expression of the L1R Gene of Vaccinia Virus by the tPA Signal Sequence Inserted in a Fowlpox-Based Recombinant Vaccine
by Antonia Radaelli, Carlo Zanotto, Chiara Brambilla, Tommaso Adami and Carlo De Giuli Morghen
Vaccines 2024, 12(10), 1115; https://doi.org/10.3390/vaccines12101115 - 29 Sep 2024
Cited by 1 | Viewed by 1465
Abstract
The use of Vaccinia virus (VACV) as a preventive vaccine against variola, the etiological agent of smallpox, led to the eradication of smallpox as a human disease. The L1 protein, a myristylated transmembrane protein present on the surface of mature virions, plays a [...] Read more.
The use of Vaccinia virus (VACV) as a preventive vaccine against variola, the etiological agent of smallpox, led to the eradication of smallpox as a human disease. The L1 protein, a myristylated transmembrane protein present on the surface of mature virions, plays a significant role in infection and morphogenesis, is well-conserved in all orthopoxviruses, and is the target of neutralizing antibodies. DNA recombinant vaccines expressing this protein were successfully used, but they showed lower efficacy in non-human and human primates when used alone, and viral-vectored fowlpox vaccines were already proved to increase immunogenicity when used as a boost. Here, we constructed a novel fowlpox-based recombinant (FPtPA-L1R), in which the tissue plasminogen activator signal sequence was linked to the 5′ end of the L1R gene to drive the L1 protein into the cellular secretion pathway. FPtPA-L1R expresses a functional heterologous protein that can be immunoprecipitated by hyperimmune rabbit serum. The protein shows cytoplasmic and membrane subcellular localizations and long-lasting expression in CEF, non-human primate Vero and human MRC-5 cells. The tissue plasminogen activator signal sequence can thus contribute significantly to the expression of the L1 protein and may enhance the immunogenicity of a putative DNA/FP prime–boost vaccine. Full article
Show Figures

Figure 1

21 pages, 4894 KiB  
Article
Development of a Two-Component Nanoparticle Vaccine Displaying an HIV-1 Envelope Glycoprotein that Elicits Tier 2 Neutralising Antibodies
by Kegomoditswe Malebo, Jeremy Woodward, Phindile Ximba, Qiniso Mkhize, Sanele Cingo, Thandeka Moyo-Gwete, Penny L. Moore, Anna-Lise Williamson and Rosamund Chapman
Vaccines 2024, 12(9), 1063; https://doi.org/10.3390/vaccines12091063 - 18 Sep 2024
Cited by 1 | Viewed by 2448
Abstract
Despite treatment and other interventions, an effective prophylactic HIV vaccine is still an essential goal in the control of HIV. Inducing robust and long-lasting antibody responses is one of the main targets of an HIV vaccine. The delivery of HIV envelope glycoproteins (Env) [...] Read more.
Despite treatment and other interventions, an effective prophylactic HIV vaccine is still an essential goal in the control of HIV. Inducing robust and long-lasting antibody responses is one of the main targets of an HIV vaccine. The delivery of HIV envelope glycoproteins (Env) using nanoparticle (NP) platforms has been shown to elicit better immunogenicity than soluble HIV Env. In this paper, we describe the development of a nanoparticle-based vaccine decorated with HIV Env using the SpyCatcher/SpyTag system. The Env utilised in this study, CAP255, was derived from a transmitted founder virus isolated from a patient who developed broadly neutralising antibodies. Negative stain and cryo-electron microscopy analyses confirmed the assembly and stability of the mi3 into uniform icosahedral NPs surrounded by regularly spaced CAP255 gp140 Env trimers. A three-dimensional reconstruction of CAP255 gp140 SpyTag–SpyCatcher mi3 clearly showed Env trimers projecting from the centre of each of the pentagonal dodecahedral faces of the NP. To our knowledge, this is the first study to report the formation of SpyCatcher pentamers on the dodecahedral faces of mi3 NPs. To investigate the immunogenicity, rabbits were primed with two doses of DNA vaccines expressing the CAP255 gp150 and a mosaic subtype C Gag and boosted with three doses of the NP-developed autologous Tier 2 CAP255 neutralising antibodies (Nabs) and low levels of heterologous CAP256SU NAbs. Full article
Show Figures

Figure 1

24 pages, 4328 KiB  
Article
Nonreciprocity in CHIKV and MAYV Vaccine-Elicited Protection
by Whitney C. Weber, Takeshi F. Andoh, Craig N. Kreklywich, Zachary J. Streblow, Michael Denton, Magdalene M. Streblow, John M. Powers, Gauthami Sulgey, Samuel Medica, Igor Dmitriev, David T. Curiel, Nicole N. Haese and Daniel N. Streblow
Vaccines 2024, 12(9), 970; https://doi.org/10.3390/vaccines12090970 - 27 Aug 2024
Cited by 1 | Viewed by 1986
Abstract
Chikungunya virus (CHIKV) is a pathogenic arthritogenic alphavirus responsible for large-scale human epidemics for which a vaccine was recently approved for use. Mayaro virus (MAYV) is a related emerging alphavirus with epidemic potential with circulation overlap potential with CHIKV. We previously reported the [...] Read more.
Chikungunya virus (CHIKV) is a pathogenic arthritogenic alphavirus responsible for large-scale human epidemics for which a vaccine was recently approved for use. Mayaro virus (MAYV) is a related emerging alphavirus with epidemic potential with circulation overlap potential with CHIKV. We previously reported the ability of a non-replicating human adenovirus (AdV)-vectored vaccine expressing the MAYV structural polyprotein to protect against disease in mice following challenge with MAYV, CHIKV and UNAV. Herein, we evaluated mouse immunity and protective efficacy for an AdV-CHIKV full structural polyprotein vaccine in combination with heterologous AdV-MAYV prime/boost regimens versus vaccine coadministration. Heterologous prime/boost regimens skewed immunity toward the prime vaccine antigen but allowed for a boost of cross-neutralizing antibodies, while vaccine co-administration elicited robust, balanced responses capable of boosting. All immunization strategies protected against disease from homologous virus infection, but reciprocal protective immunity differences were revealed upon challenge with heterologous viruses. In vivo passive transfer experiments reproduced the inequity in reciprocal cross-protection after heterologous MAYV challenge. We detected in vitro antibody-dependent enhancement of MAYV replication, suggesting a potential mechanism for the lack of cross-protection. Our findings provide important insights into rational alphavirus vaccine design that may have important implications for the evolving alphavirus vaccine landscape. Full article
(This article belongs to the Section Vaccines against Tropical and other Infectious Diseases)
Show Figures

Figure 1

15 pages, 4981 KiB  
Article
Sequential Immunization with Vaccines Based on SARS-CoV-2 Virus-like Particles Induces Broadly Neutralizing Antibodies
by Youjun Mi, Kun Xu, Wenting Wang, Weize Kong, Xiaonan Xu, Xifeng Rong and Jiying Tan
Vaccines 2024, 12(8), 927; https://doi.org/10.3390/vaccines12080927 - 19 Aug 2024
Viewed by 1646
Abstract
Although many people have been vaccinated against COVID-19, infections with SARS-CoV-2 seem hard to avoid. There is a need to develop more effective vaccines and immunization strategies against emerging variants of infectious diseases. To understand whether different immunization strategies using variants sequence-based virus-like [...] Read more.
Although many people have been vaccinated against COVID-19, infections with SARS-CoV-2 seem hard to avoid. There is a need to develop more effective vaccines and immunization strategies against emerging variants of infectious diseases. To understand whether different immunization strategies using variants sequence-based virus-like particles (VLPs) vaccines could offer superior immunity against future SARS-CoV-2 variants, our team constructed VLPs for the original Wuhan-Hu-1 strain (prototype), Delta (δ) variant, and Omicron (ο) variant of SARS-CoV-2, using baculovirus-insect expression system. Then we used these VLPs to assess the immune responses induced by homologous prime-boost, heterologous prime-boost, and sequential immunizations strategies in a mouse model. Our results showed that the pro+δ+ο sequential strategies elicited better neutralizing antibody responses. These sequential strategies also take advantage of inducing CD4+ T and CD8+ T lymphocytes proliferation and tendency to cytokine of Th1. Currently, our data suggest that sequential immunization with VLPs of encoding spike protein derived from SARS-CoV-2 variants of concern may be a potential vaccine strategy against emerging diseases, such as “Disease X”. Full article
(This article belongs to the Special Issue SARS-CoV-2 Infections; Treatment and Development of Vaccine)
Show Figures

Figure 1

21 pages, 3003 KiB  
Article
Characterization of the Anti-Viral and Vaccine-Specific CD8+ T Cell Composition upon Treatment with the Cancer Vaccine VSV-GP
by Tamara Hofer, Lisa Pipperger, Sarah Danklmaier, Krishna Das and Guido Wollmann
Vaccines 2024, 12(8), 867; https://doi.org/10.3390/vaccines12080867 - 1 Aug 2024
Viewed by 2078
Abstract
Numerous factors influence the magnitude and effector phenotype of vaccine-induced CD8+ T cells, thereby potentially impacting treatment efficacy. Here, we investigate the effect of vaccination dose, route of immunization, presence of a target antigen-expressing tumor, and heterologous prime-boost with peptide vaccine partner [...] Read more.
Numerous factors influence the magnitude and effector phenotype of vaccine-induced CD8+ T cells, thereby potentially impacting treatment efficacy. Here, we investigate the effect of vaccination dose, route of immunization, presence of a target antigen-expressing tumor, and heterologous prime-boost with peptide vaccine partner following vaccination with antigen-armed VSV-GP. Our results indicate that a higher vaccine dose increases antigen-specific CD8+ T cell proportions while altering the phenotype. The intravenous route induces the highest proportion of antigen-specific CD8+ T cells together with the lowest anti-viral response followed by the intraperitoneal, intramuscular, and subcutaneous routes. Moreover, the presence of a B16-OVA tumor serves as pre-prime, thereby increasing OVA-specific CD8+ T cells upon vaccination and thus altering the ratio of anti-tumor versus anti-viral CD8+ T cells. Interestingly, tumor-specific CD8+ T cells exhibit a different phenotype compared to bystander anti-viral CD8+ T cells. Finally, the heterologous combination of peptide and viral vaccine elicits the highest proportion of antigen-specific CD8+ T cells in the tumor and tumor-draining lymph nodes. In summary, we provide a basic immune characterization of various factors that affect anti-viral and vaccine target-specific CD8+ T cell proportions and phenotypes, thereby enhancing our vaccinology knowledge for future vaccine regimen designs. Full article
Show Figures

Figure 1

16 pages, 2603 KiB  
Article
Bimodal Effect of NKG2A Blockade on Intratumoral and Systemic CD8 T Cell Response Induced by Cancer Vaccine
by Erika Riva, Susanna Carboni, Wilma di Berardino-Besson, Mati Moyat, Elodie Belnoue, Laetitia Devy-Dimanche and Matteo Rossi
Cancers 2024, 16(11), 2036; https://doi.org/10.3390/cancers16112036 - 27 May 2024
Viewed by 1684
Abstract
Immune check-point blockade (ICB) has revitalized cancer immunotherapy, showing unprecedented efficacy despite only a narrow number of indications and with limited long-term protection. Cancer vaccines are promising combination partners for ICB to widen the patient population profiting from these treatments. Therapeutic heterologous prime-boost [...] Read more.
Immune check-point blockade (ICB) has revitalized cancer immunotherapy, showing unprecedented efficacy despite only a narrow number of indications and with limited long-term protection. Cancer vaccines are promising combination partners for ICB to widen the patient population profiting from these treatments. Therapeutic heterologous prime-boost vaccination with KISIMATM protein vaccine and VSV-GP-TAg oncolytic virus was shown to inflame the tumor microenvironment, promoting significant infiltration of antigen-specific CD8 T cells resulting in robust antitumoral efficacy in mouse tumor models, and clinical trials are currently ongoing. Here, we report the impact of NKG2A blockade on antitumoral CD8 T cell immune response elicited by KISIMA—VSV-GP-TAg vaccination in tumor mouse models. Combination therapy significantly reduced the amount of vaccine-induced exhausted CD8 T cells infiltrating the tumor, resulting in short-term improved tumor growth control and prolonged mouse survival, while it also influenced the establishment of systemic effector memory CD8 T cell response. Taken together, these data show a compartment-dependent effect of NKG2A blockade on cancer vaccine-induced T cell immunity, increasing intratumoral T cell efficacy and attenuating the development of peripheral effector memory CD8 T cell response. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Back to TopTop