Добірка наукової літератури з теми "Heterologous prime-boost"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся зі списками актуальних статей, книг, дисертацій, тез та інших наукових джерел на тему "Heterologous prime-boost".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Статті в журналах з теми "Heterologous prime-boost"

1

Lu, Shan. "Heterologous prime–boost vaccination." Current Opinion in Immunology 21, no. 3 (June 2009): 346–51. http://dx.doi.org/10.1016/j.coi.2009.05.016.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Pan, Chien-Hsiung, Hui-mei Hu, Yu-Ju Hsiao, and Sze-Hsien Wu. "Heterologous prime-boost vaccination with DNA vaccine and recombinant subunit containing four serotypes of dengue virus envelope protein domain III elicits neutralizing antibodies and specific T-cell responses (P4327)." Journal of Immunology 190, no. 1_Supplement (May 1, 2013): 123.27. http://dx.doi.org/10.4049/jimmunol.190.supp.123.27.

Повний текст джерела
Анотація:
Abstract Dengue is still an important public health problem and no dengue vaccine is available now. In this study, we characterized prime-boost vaccine regimens using heterologous and homologous dengue DNA vaccine and recombinant subunit vaccine consisted of envelope (E) protein domain III (ED III) from four serotypes of dengue virus (DENV). A broader and balanced IFN-gamma and IL-4 responses was observed in DNA-prime and subunit-boost immunized mice, compared to a narrower but strong IFN-gamma production after homologous DNA vaccination and an IL-4 only response in homologous subunit vaccine immunized mice. In addition, a significant ED III specific antibody response was detected in all immunized mice except vector control. The highest IgG titer was appeared in homologous subunit vaccine immunized mice, followed by heterologous prime-boost regimen and homologous DNA vaccine. However, that heterologous prime-boost immunized mice generated a highest neutralizing antibody than the others, suggested a better protection provided by this DNA-prime and subunit vaccine-boost regimen.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Tang, Xian, and Zhiwei Chen. "Heterologous immunization induces potent anti-SIV immunity with qualitative and quantitative improvements (53.22)." Journal of Immunology 186, no. 1_Supplement (April 1, 2011): 53.22. http://dx.doi.org/10.4049/jimmunol.186.supp.53.22.

Повний текст джерела
Анотація:
Abstract Background: The live replication-competent modified vaccinia virus Tiantan (MVTT) is an attractive vaccine vector. In this study, we constructed a recombinant MVTT expressing SIV Gag-Pol and Env (MVTT-SIV) and compared its efficacy for inducing antigen-specific immunity when administrated in different routes with or without rAd5-based vaccine encoding the same antigens (rAd5-SIV). Methods: MVTT-SIV was tested for its immunogenicity in mice administered homologously by a prime-boost strategy or heterologously in combination with rAd5-SIV. The prime routes included intranasal (IN), sublingual (SL), oral, mixed (mix of the previous three musocal routes) or intramuscular (IM) injection. All groups received IM boost. Result: Mice given heterologous boost produced significantly greater SIV specific T cell and Gag-specific antibody responses than homologous boost groups. Besides, heterologous immunization generated much higher level of neutralizing antibodies, despite of the lower titers of Env-specific binding antibodies according to Elisa assay. Moreover, the immunogenicity of MVTT-SIV given by mixed route followed by rAd5-SIV intramuscular boost was unique in terms of anti-SIV cellular and humoral immunity. Conclusion: Heterologous immunization induces potent anti-SIV Immunity with improvements in quality and quantity. The combination of MVTT-SIV mixed prime and rAd5-SIV intramuscular boost may be an effective and protective vaccination strategy in further studies.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

ZHANG, G., V. T. T. HUONG, B. BATTUR, J. ZHOU, H. ZHANG, M. LIAO, O. KAWASE, et al. "A heterologous prime-boost vaccination regime using DNA and a vaccinia virus, both expressing GRA4, induced protective immunity againstToxoplasma gondiiinfection in mice." Parasitology 134, no. 10 (May 17, 2007): 1339–46. http://dx.doi.org/10.1017/s0031182007002892.

Повний текст джерела
Анотація:
SUMMARYThe dense granule antigen 4 (GRA4) is known as an immundominant antigen ofToxoplasma gondiiand, therefore, is considered as a vaccine candidate. For further evaluation of its vaccine effect, a recombinant plasmid and vaccinia virus, both expressing GRA4, were constructed, and a heterologous prime-boost vaccination regime was performed in a mouse model. The mice immunized with the heterologous prime-boost vaccination regime showed a high level of specific antibody response against GRA4 and a significantly high level of gamma interferon (IFN-γ) production and survived completely against a subsequent challenge infection with a lethal dose ofT. gondii. In addition, the formation of cysts was inhibited in the mice vaccinated with the heterologous prime-boost vaccination regime. These results demonstrate that the heterologous prime-boost vaccination regime using DNA and a vaccinia virus, both expressing GRA4, could induce both humoral and cellular immune responses and provide effective protection against lethal acute and chronicT. gondiiinfections in mice.
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Garg, Ishan, Abu Baker Sheikh, Suman Pal, and Rahul Shekhar. "Mix-and-Match COVID-19 Vaccinations (Heterologous Boost): A Review." Infectious Disease Reports 14, no. 4 (July 20, 2022): 537–46. http://dx.doi.org/10.3390/idr14040057.

Повний текст джерела
Анотація:
Various safe and effective COVID-19 vaccines utilizing different platforms (mRNA, adenovirus vector, inactivated virus-based) are available against SARS-CoV-2 infection. A prime-boost regimen (administration of two doses) is recommended to induce an adequate and sustained immune response. Most of these vaccines follow a homologous regimen (the same type of vaccine as priming and booster doses). However, there is a growing interest in a heterologous prime-boost vaccination regimen to potentially help address concerns posed by fluctuating vaccine supplies, serious adverse effects (anaphylaxis and thromboembolic episodes following adenovirus-based vaccines), new emerging virulent strains, inadequate immune response in immunocompromised individuals, and waning immunity. Various studies have demonstrated that heterologous prime-boost vaccination may induce comparable or higher antibody (spike protein) titers and a similar reactogenicity profile to the homologous prime-boost regimen. Based on these considerations, the Center for Disease Control and Prevention has issued guidance supporting the “mix-and-match” heterologous boost COVID-19 vaccine strategy.
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Folegatti, Pedro M., Amy Flaxman, Daniel Jenkin, Rebecca Makinson, Lucy Kingham-Page, Duncan Bellamy, Fernando Ramos Lopez, et al. "Safety and Immunogenicity of Adenovirus and Poxvirus Vectored Vaccines against a Mycobacterium Avium Complex Subspecies." Vaccines 9, no. 3 (March 16, 2021): 262. http://dx.doi.org/10.3390/vaccines9030262.

Повний текст джерела
Анотація:
Heterologous prime-boost strategies are known to substantially increase immune responses in viral vectored vaccines. Here we report on safety and immunogenicity of the poxvirus Modified Vaccinia Ankara (MVA) vectored vaccine expressing four Mycobacterium avium subspecies paratuberculosis antigens as a single dose or as a booster vaccine following a simian adenovirus (ChAdOx2) prime. We demonstrate that a heterologous prime-boost schedule is well tolerated and induced T-cell immune responses.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Carlétti, Dyego, Denise Morais da Fonseca, Ana Flávia Gembre, Ana Paula Masson, Lívia Weijenborg Campos, Luciana C. C. Leite, Andréa Rodrigues Pires, et al. "A Single Dose of a DNA Vaccine Encoding Apa Coencapsulated with 6,6′-Trehalose Dimycolate in Microspheres Confers Long-Term Protection against Tuberculosis in Mycobacterium bovis BCG-Primed Mice." Clinical and Vaccine Immunology 20, no. 8 (June 5, 2013): 1162–69. http://dx.doi.org/10.1128/cvi.00148-13.

Повний текст джерела
Анотація:
ABSTRACTMycobacterium bovisBCG prime DNA (Mycobacterium tuberculosisgenes)-booster vaccinations have been shown to induce greater protection against tuberculosis (TB) than BCG alone. This heterologous prime-boost strategy is perhaps the most realistic vaccination for the future of TB infection control, especially in countries where TB is endemic. Moreover, a prime-boost regimen using biodegradable microspheres seems to be a promising immunization to stimulate a long-lasting immune response. The alanine proline antigen (Apa) is a highly immunogenic glycoprotein secreted byM. tuberculosis. This study investigated the immune protection of Apa DNA vaccine against intratrachealM. tuberculosischallenge in mice on the basis of a heterologous prime-boost regimen. BALB/c mice were subcutaneously primed with BCG and intramuscularly boosted with a single dose of plasmid carryingapaand 6,6′-trehalose dimycolate (TDM) adjuvant, coencapsulated in microspheres (BCG-APA), and were evaluated 30 and 70 days after challenge. This prime-boost strategy (BCG-APA) resulted in a significant reduction in the bacterial load in the lungs, thus leading to better preservation of the lung parenchyma, 70 days postinfection compared to BCG vaccinated mice. The profound effect of this heterologous prime-boost regimen in the experimental model supports its development as a feasible strategy for prevention of TB.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Dai, Ming-Shen, Ren-In You, Yu-Feng Hsieh, Kai-Yu Lo, Huey-Kang Sytwu, Georges Vassaux, and Tsu-Yi Chao. "Early Treg suppression by a Listeriolysin-O-expressing E. coli vaccine in heterologous prime-boost vaccination against cancer (165.49)." Journal of Immunology 186, no. 1_Supplement (April 1, 2011): 165.49. http://dx.doi.org/10.4049/jimmunol.186.supp.165.49.

Повний текст джерела
Анотація:
Abstract [Background] Earlier studies have shown that higher CD8+ T-cell response and better tumor protection can be achieved by heterologous prime-boost vaccination in mice. We previously demonstrated that a Listeriolysin-O (LLO)-expressing E. coli vaccine can enhanced CD8-cytotoxic T cell (CTL) responses by mitigating regulatory T cells (Treg)-mediated suppression. We herein employed this Treg-inhibiting vaccine into the prime/boost immunization strategy. [Methods] Bacteria E. coli-LLO expressing Ovalbumin (OVA) and plasmid pcDNA—encoding OVA were used as specific antigen immunization. C57B6 mice and syngenic melanoma cell line B16-OVA were used in tumor challenge model. CD25 monoclonal antibody (PC-61) was used for Treg depletion. [Results] Higher OVA-specific CD8+ T-cell responses and superior tumor inhibition were seen in the bacterial-prime/plasmid-boost setting than homologous or reversed sequence. This tumor protection effect from heterologous prime/boost remained significant in the therapeutic model. When examining the Treg effect during the prime/boost immunization, we found that only early Treg-suppression/depletion could lead to better antigen-specific CTL and tumor response. [Conclusion] Our studies offer the first evidence that a Listeriolysin-O E. coli vaccine can induce superior anti-tumor effect in conjunction with DNA in a heterologous prime-boost protocol and proposed that early Treg inhibition is crucial to a successful immunization against cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Santra, Sampa, Yue Sun, Jenny G. Parvani, Valerie Philippon, Michael S. Wyand, Kelledy Manson, Alicia Gomez-Yafal, et al. "Heterologous Prime/Boost Immunization of Rhesus Monkeys by Using Diverse Poxvirus Vectors." Journal of Virology 81, no. 16 (June 6, 2007): 8563–70. http://dx.doi.org/10.1128/jvi.00744-07.

Повний текст джерела
Анотація:
ABSTRACT As the diversity of potential immunogens increases within certain classes of vectors, the possibility has arisen of employing heterologous prime/boost immunizations using diverse members of the same family of vectors. The present study was initiated to explore the use of divergent pox vectors in a prime/boost regimen to elicit high-frequency cellular immune responses to human immunodeficiency virus type 1 envelope and simian immunodeficiency virus gag in rhesus monkeys. We demonstrated that monkeys vaccinated with a recombinant modified vaccinia virus Ankara (rMVA) prime/recombinant fowlpox virus (rFPV) boost regimen and monkeys vaccinated with a recombinant vaccinia virus prime/rFPV boost regimen developed comparable cellular immune responses that were greater in magnitude than those elicited by a homologous prime/boost with rMVA. Nevertheless, comparable magnitude recall cellular immune responses were observed in monkeys vaccinated with heterologous and homologous recombinant poxvirus following challenge with the CXCR4-tropic SHIV-89.6P. Consistent with this finding, comparable levels of containment of viral replication and CD4+ T-lymphocyte preservation were seen in these groups of recombinant poxvirus-vaccinated monkeys. This study supports further exploration of combining recombinant vectors of the same family in prime/boost immunization strategies to optimize vaccine-elicited cellular immune responses.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Kaku, Chengzi I., Elizabeth R. Champney, Johan Normark, Marina Garcia, Carl E. Johnson, Clas Ahlm, Wanda Christ, et al. "Broad anti–SARS-CoV-2 antibody immunity induced by heterologous ChAdOx1/mRNA-1273 vaccination." Science 375, no. 6584 (March 4, 2022): 1041–47. http://dx.doi.org/10.1126/science.abn2688.

Повний текст джерела
Анотація:
Heterologous prime-boost immunization strategies have the potential to augment COVID-19 vaccine efficacy. We longitudinally profiled severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S)–specific serological and memory B cell (MBC) responses in individuals who received either homologous (ChAdOx1:ChAdOx1) or heterologous (ChAdOx1:mRNA-1273) prime-boost vaccination. Heterologous messenger RNA (mRNA) booster immunization induced higher serum neutralizing antibody and MBC responses against SARS-CoV-2 variants of concern (VOCs) compared with that of homologous ChAdOx1 boosting. Specificity mapping of circulating B cells revealed that mRNA-1273 boost immunofocused ChAdOx1-primed responses onto epitopes expressed on prefusion-stabilized S. Monoclonal antibodies isolated from mRNA-1273–boosted participants displayed overall higher binding affinities and increased breadth of reactivity against VOCs relative to those isolated from ChAdOx1-boosted individuals. Overall, the results provide molecular insight into the enhanced quality of the B cell response induced after heterologous mRNA booster vaccination.
Стилі APA, Harvard, Vancouver, ISO та ін.

Дисертації з теми "Heterologous prime-boost"

1

Badamchi-Zadeh, Alexander. "Heterologous prime-boost vaccine regimens against Chlamydia trachomatis." Thesis, Imperial College London, 2014. http://hdl.handle.net/10044/1/25021.

Повний текст джерела
Анотація:
Chlamydia trachomatis is the most common bacterial sexually transmitted disease in man and despite decades of effort, there is still no protective vaccine. Left untreated, genital infection can lead to pelvic inflammatory disease, ectopic pregnancy, and infertility. However, infection-induced immunity in both animal models and humans indicates a strong role for CD4+ Th1-biased immune responses. Using a multi-component vaccine approach we assessed the immunogenicity and protective efficacy of novel plasmid DNA, Adenovirus 5 (HuAd5) and modified vaccinia Ankara (MVA) vectors each containing a major outer membrane protein (MOMP) transgene and recombinant MOMP protein in various heterologous prime-boost regimens in BALB/c and B6C3F1 mice. During the course of the prime-boost regimens, serum and vaginal MOMP-specific antibody titres, subtypes, avidities and neutralisation abilities were assessed, alongside IFN-γ ELISpot and CD4+ and CD8+ T cell polyfunctionality (IFN-γ, TNF-α, and IL-2). Regimens were grouped on the distinct MOMP-specific immune environments they elicited, with these regimens taken through into C. trachomatis vaginal challenge studies in two mouse models to shed light on the relative contribution of each environment to protective immunity. The DNA-HuAd5-MVA-Protein (D.A.M.P.) vaccine regimen resulted in a significant reduction in C. trachomatis vaginal shedding at day 3 post-infection in both BALB/c and B6C3F1 mouse strains. This significant reduction was lost when D.A.M.P. vaccinated mice were depleted of their CD4+ T cells prior to challenge, indicating the protection is CD4+ T cell mediated. C. trachomatis EB serum neutralisation profiles were similar between protective and non-protective vaccine regimens and combined with passive transfer experiments into naïve C57BL/6 mice and IFN-γ knock-out C57BL/6 mice we concluded that the antibody response did not play a significant role in this vaccine-induced protection. As well as infecting the genital tract, Chlamydia trachomatis is also the causal agent of trachoma, the leading cause of infectious blindness in the world. Recently, Kari et al. revealed serum anti-MOMP antibodies correlated with the reduction in chlamydial ocular burden in non-human primates, while anti-PmpD and anti-Pgp3 serum antibodies correlated with chlamydial eradication. We therefore investigated if we could induce such anti-chlamydial antibodies on the murine eye through heterologous prime-boost vaccinations. We uncovered a vaccination regimen that induced significantly greater anti-MOMP ocular antibodies, and employed this regimen for the additional chlamydial antigens, of which all induced ocular antigen-specific IgG antibodies. This is the first investigation into such vaccination regimens to induce chlamydial specific ocular antibodies and provides a new model for the screening of future potential trachoma vaccines.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Lange, Uta Gisela. "Heterologous prime-boost strategies in the development of novel vaccines against leishmaniasis." Thesis, University of Cambridge, 2003. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.619876.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Philosof, Bar. "A bacterium from the human microbiota as a vaccine vector. Efficient priming of the murine immune system by vaginal delivery of recombinant Streptococcus gordonii." Doctoral thesis, Università di Siena, 2022. http://hdl.handle.net/11365/1216735.

Повний текст джерела
Анотація:
The ability to prime the immune system against an antigen, and to rapidly recall this response upon antigen reencounter is a fundamental characteristic of the adaptive immune response. The association of an antigen recognized by the immune system in a certain tissue, with the same antigen encountered at a later timepoint in a different tissue, is of primary importance to obtain a systemic and effective immune response and is the foundation behind the utilization of vaccines. The study of vaccine delivery platforms that may activate the immune system in such a manner is therefore of primary importance in the effort to design novel vaccine formulation and prime-boost strategies. The aim of the present work was to study the in vivo priming effect induced by a recombinant Streptococcus gordonii vaccine vector expressing a heterologous antigen and delivered to the vaginal tract, a unique mucosal tissue. To study the priming effect induced by the recombinant Streptococcus gordonii, we employed a prime-boost strategy and compared the cellular an humoral immune response towards the soluble antigen between recombinant- and WT-immunized mice. Using this model, we show that vaginal immunization with the recombinant Streptococcus gordonii elicited systemic production of antigen-specific antibodies, shifted the IgG subclasses profile, led to an increase in plasma cells in the lymph nodes, a higher number of antigen-specific antibody-secreting cells in the spleen and modulated the cytokine expression profile of splenocytes. The longevity of the priming effect induced by the recombinant vector was also analyzed by comparing three and six months boosting schedules. We found that the priming is boostable with a similar efficacy for at least six months (Chapter 3). These data demonstrate that vaginal priming with the recombinant S. gordonii vector results in a systemic activation of both cellular and humoral immune compartments, and that this priming effect is long-lived without significant immune waning. In this study, we also assessed the transcriptomic response of splenocytes from S. gordonii-immunized mice towards the antigen. We observed differences in immune pathways between recombinant- and WT-immunized mice, and also between the three- and six-months boosted groups (Chapter 4). In addition, we observed a gene signature correlated with antigen-specific IgG titers. These findings suggest that the immune system’s encounter with the antigen on the surface of the recombinant S. gordonii in the vaginal tract results in a differential immune activation in in response to the antigen. This work contributes to the knowledge on the capability of recombinant live vaccine vectors delivered mucosally to prime and modulate the immune response, and has important implications in the design of innovative vaccination strategies.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Searle, Benjamin James. "Characterisation of heterologous prime-boost vaccination strategies : an investigation into the nature and delivery of vaccines and the subsequent generation of immune responses." Thesis, University of Glasgow, 2004. http://theses.gla.ac.uk/2779/.

Повний текст джерела
Анотація:
This thesis describes work undertaken to consider how strategies of vaccination can be manipulated to generate specific types and magnitude of immune response to prevent infection or treat established infection. The vaccination strategies employed were based on the initial delivery of DNA vaccine through intramuscular injection or ballistic delivery using a gene gun, followed by heterologous boosts, based on the same antigen delivered using an alternative route. These boost strategies included a) intramuscular delivery of purified recombinant HBcAg, b) mucosal delivery of HBcAg expressed by an attenuated strain of Salmonella typhimurium, and c) intranasal delivery of the protein accompanied by a mucosal adjuvant. Following each vaccination regime tested a number of specific immune responses, including serum and mucosal antibody production, CD4+ T helper proliferation in spleens and lymph nodes, CD8+ T cell activation and killing were measured. These experiments revealed that the character of the immune responses primed in response to DNA vaccination differed according to route of immunisation, with intramuscular vaccination inducing a rapid CTL response. CD4+ T cells generated appeared to be of the Th1 phenotype and showed the strongest localisation in the spleen. In contrast, following vaccination with the gene gun, CD4+ cells of a Th2 phenotype were generated with responses being found to be stronger in the local draining lymph nodes that the spleen. Combining DNA delivered using the intramuscular route with recombinant purified protein delivered by the same route was effective at boosting systemic antibody titres, as was boosting using attenuated Salmonella expressing the same antigen when delivered to the mucosal surface of the gut. In contrast, boosting of DNA primed animals with purified protein, even in the presence of an appropriate mucosal adjuvant, was not successful at increasing titres of systemic antibodies. Interestingly, specific mucosal immune responses were absent when either of the heterologous vaccination strategies used a mucosal boosting approach.
Стилі APA, Harvard, Vancouver, ISO та ін.
5

"Plague vaccine: Homologous and heterologous prime-boost strategies using recombinant Yersinia pestis proteins." Tulane University, 2005.

Знайти повний текст джерела
Анотація:
Yersinia pestis has been identified as a potential agent of biological warfare or bioterrorism and presents a particular challenge to vaccine developers because of the severity and potential transmissibility of the pneumonic form of the disease in humans. A Y. pestis-derived fusion protein (F1-V) has shown great promise as a protective antigen in murine studies. Here, initial studies were done to examine the ability of different prime-boost regimens, including parenteral, mucosal, and transcutaneous delivery, to induce a specific immune response of high titer and long duration. Next, we examine different prime-boost regimens in order to determine if heterologous boosting can provide protection against aerosol challenge when compared to homologous boosting. Additionally, we examine whether non-parenteral immunization can prime for a parenteral boost. Finally, we examine the role of the adjuvant in inducing a protective immune response. The most significant findings of the studies reported here are that (1) heterologous boosting can be as or more effective than homologous boosting for induction of either serum or BAL anti-F1-V IgG1 responses, (2) heterologous boosting protects mice as well as homologous boosting against aerosol challenge with Y. pestis , (3) parenteral immunization is not required to protect mice against aerosolized plague challenge, (4) the route of immunization and choice of adjuvant influence the magnitude of the antibody response as well as the IgG1/IgG2a ratio, and (5) inclusion of an appropriate adjuvant is critical for non-parenteral immunization
acase@tulane.edu
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Huang, Yu-Hsuan, and 黃于璇. "Enhanced immune responses elicited by heterologous prime-boost vaccination using DNA/adenoviral vectors and protein of H5N1 hemagglutinin." Thesis, 2010. http://ndltd.ncl.edu.tw/handle/92699131900412448191.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

"Searching for an HIV Vaccine: A Heterologous Prime-boost System using Replicating Vaccinia Virus and Plant-produced Virus-like Particles." Doctoral diss., 2016. http://hdl.handle.net/2286/R.I.40210.

Повний текст джерела
Анотація:
abstract: The HIV-1 pandemic continues to cause millions of new infections and AIDS-related deaths each year, and a majority of these occur in regions of the world with limited access to antiretroviral therapy. Therefore, an HIV-1 vaccine is still desperately needed. The most successful HIV-1 clinical trial to date used a non-replicating canarypox viral vector and protein boosting, yet its modest efficacy left room for improvement. Efforts to derive novel vectors which can be both safe and immunogenic, have spawned a new era of live, viral vectors. One such vaccinia virus vector, NYVAC-KC, was specifically designed to replicate in humans and had several immune modulators deleted to improve immunogenicity and reduce pathogenicity. Two NYVAC-KC vectors were generated: one expressing the Gag capsid, and one with deconstructed-gp41 (dgp41), which contains an important neutralizing antibody target, the membrane proximal external region (MPER). These vectors were combined with HIV-1 Gag/dgp41 virus-like particles (VLPs) produced in the tobacco-relative Nicotiana benthamiana. Different plant expression vectors were compared in an effort to improve yield. A Geminivirus-based vector was shown to increase the amount of MPER present in VLPs, thus potentially enhancing immunogenicity. Furthermore, these VLPs were shown to interact with the innate immune system through Toll-like receptor (TLR) signaling, which activated antigen presenting cells to induce a Th2-biased response in a TLR-dependent manner. Furthermore, expression of Gag and dgp41 in NYVAC-KC vectors resulted in activation of antiviral signaling pathways reliant on TBK1/IRF3, which necessitated the use of higher doses in mice to match the immunogenicity of wild-type viral vectors. VLPs and NYVAC-KC vectors were tested in mice, ultimately showing that the best antibody and Gag-specific T cell responses were generated when both components were administered simultaneously. Thus, plant-produced VLPs and poxvirus vectors represent a highly immunogenic HIV-1 vaccine candidate that warrants further study.
Dissertation/Thesis
Doctoral Dissertation Biological Design 2016
Стилі APA, Harvard, Vancouver, ISO та ін.

Частини книг з теми "Heterologous prime-boost"

1

Palmowski, Michael J., and Caroline Smith. "Heterologous Prime-Boost Vaccination in Tumor Immunotherapy." In Handbook of Cancer Vaccines, 115–20. Totowa, NJ: Humana Press, 2004. http://dx.doi.org/10.1007/978-1-59259-680-5_9.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Jackson, Ronald J., David B. Boyle, and Charani Ranasinghe. "Progresses in DNA-Based Heterologous Prime-Boost Immunization Strategies." In Methods in Molecular Biology, 61–90. New York, NY: Springer New York, 2014. http://dx.doi.org/10.1007/978-1-4939-0410-5_5.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

McShane, Helen, Adrian Hill, and Andrew McMichael. "Overview of Heterologous Prime-Boost Immunization Strategies." In New Generation Vaccines, Fourth Edition, 396–404. CRC Press, 2009. http://dx.doi.org/10.3109/9781420060744-39.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Shnawa, Ibrahim M. S. "Heterologous Prime-boost as COVID-19 Vaccine Strategies: Towards a Nationwide Implementation." In Challenges and Advances in Pharmaceutical Research Vol. 4, 72–83. Book Publisher International (a part of SCIENCEDOMAIN International), 2022. http://dx.doi.org/10.9734/bpi/capr/v4/16535d.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Srivastava, Indresh, Jeffrey Ulmer, Margaret Liu, Adrian Hill, Joerg Schneider, and Andrew McMichael. "DNA-Modified Virus Ankara and Other Heterologous Prime-Boost Immunization Strategies for Effector T Cell Induction." In New Generation Vaccines, Fourth Edition. Informa Healthcare, 2004. http://dx.doi.org/10.1201/9781439834404.ch31.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

"DNA-Modified Virus Ankara and Other Heterologus Prime-Boost Immunization Strategies for Effector T Cell Induction." In New Generation Vaccines, 769–94. CRC Press, 2004. http://dx.doi.org/10.1201/9781439834404-38.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.

Тези доповідей конференцій з теми "Heterologous prime-boost"

1

Azevedo, Patrick, Natália Souza, Lídia Faustino, Beatriz Santos, Alexandre Machado, and Ricardo Gazzinelli. "Evaluation of humoral and cellular immune response after heterologous prime-boost immunization against SARS-CoV-2." In International Symposium on Immunobiological. Instituto de Tecnologia em Imunobiológicos, 2021. http://dx.doi.org/10.35259/isi.2021_46710.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Blair, Wade, Gijsbert Grotenbreg, Ciaran Scallan, Amy Rappaport, Renee Greer, Leonid Gitlin, Kieu Lam, et al. "Abstract 724: A novel heterologous prime boost vaccine system drives tumor specific and potent CD8 T cell responses for cancer immunotherapy." In Proceedings: AACR Annual Meeting 2018; April 14-18, 2018; Chicago, IL. American Association for Cancer Research, 2018. http://dx.doi.org/10.1158/1538-7445.am2018-724.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

KIM, SUNGBAE, KyungHae Jung, JinHee Ahn, and Jeongeun Kim. "Abstract CT414: Efficient induction of cellular and humoral immune responses by heterologous prime-boost therapeutic vaccination, involving plasmid DNA and adenoviral vector, in subjects with HER2-expressing breast cancer: Results from a phase Ib study." In Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. American Association for Cancer Research, 2014. http://dx.doi.org/10.1158/1538-7445.am2014-ct414.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії