Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Immunol ; 197(4): 1242-51, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27412417

RESUMEN

There is great interest in the development of Ab-inducing subunit vaccines targeting infections, including HIV, malaria, and Ebola. We previously reported that adenovirus vectored vaccines are potent in priming Ab responses, but uncertainty remains regarding the optimal approach for induction of humoral immune responses. In this study, using OVA as a model Ag, we assessed the magnitude of the primary and anamnestic Ag-specific IgG responses of mice to four clinically relevant vaccine formulations: replication-deficient adenovirus; modified vaccinia Ankara (a poxvirus); protein with alum; and protein in the squalene oil-in-water adjuvant Addavax. We then used flow cytometric assays capable of measuring total and Ag-specific germinal center (GC) B cell and follicular Th cell responses to compare the induction of these responses by the different formulations. We report that adenovirus vectored vaccines induce Ag insert-specific GC B cell and Ab responses of a magnitude comparable to those induced by a potent protein/squalene oil-in-water formulation whereas-despite a robust overall GC response-the insert-specific GC B cell and Ab responses induced by modified vaccinia Ankara were extremely weak. Ag-specific follicular Th cell responses to adenovirus vectored vaccines exceeded those induced by other platforms at day 7 after immunization. We found little evidence that innate immune activation by adenovirus may act as an adjuvant in such a manner that the humoral response to a recombinant protein may be enhanced by coadministering with an adenovirus lacking a transgene of interest. Overall, these studies provide further support for the use of replication-deficient adenoviruses to induce humoral responses.


Asunto(s)
Adenoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Linfocitos B/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas Virales/inmunología , Animales , Antígenos/inmunología , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Vectores Genéticos , Centro Germinal/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología
2.
Mol Ther ; 22(12): 2142-2154, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25156127

RESUMEN

The development of effective vaccines against difficult disease targets will require the identification of new subunit vaccination strategies that can induce and maintain effective immune responses in humans. Here we report on a phase 1a clinical trial using the AMA1 antigen from the blood-stage Plasmodium falciparum malaria parasite delivered either as recombinant protein formulated with Alhydrogel adjuvant with and without CPG 7909, or using recombinant vectored vaccines--chimpanzee adenovirus ChAd63 and the orthopoxvirus MVA. A variety of promising "mixed-modality" regimens were tested. All volunteers were primed with ChAd63, and then subsequently boosted with MVA and/or protein-in-adjuvant using either an 8- or 16-week prime-boost interval. We report on the safety of these regimens, as well as the T cell, B cell, and serum antibody responses. Notably, IgG antibody responses primed by ChAd63 were comparably boosted by AMA1 protein vaccine, irrespective of whether CPG 7909 was included in the Alhydrogel adjuvant. The ability to improve the potency of a relatively weak aluminium-based adjuvant in humans, by previously priming with an adenoviral vaccine vector encoding the same antigen, thus offers a novel vaccination strategy for difficult or neglected disease targets when access to more potent adjuvants is not possible.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Protozoos/administración & dosificación , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Adenovirus de los Simios/genética , Adulto , Hidróxido de Aluminio/administración & dosificación , Antígenos de Protozoos/inmunología , Terapia Combinada , Vectores Genéticos/administración & dosificación , Humanos , Inmunización Secundaria , Masculino , Persona de Mediana Edad , Oligodesoxirribonucleótidos/administración & dosificación , Orthopoxvirus/genética , Vacunación , Adulto Joven
3.
Immunology ; 141(4): 628-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24303947

RESUMEN

Acquisition of non-sterilizing natural immunity to Plasmodium falciparum malaria has been shown in low transmission areas following multiple exposures. However, conflicting data from endemic areas suggest that the parasite may interfere with the induction of effective B-cell responses. To date, the impact of blood-stage parasite exposure on antigen-specific B cells has not been reported following controlled human malaria infection (CHMI). Here we analysed human B-cell responses in a series of Phase I/IIa clinical trials, which include CHMI, using candidate virus-vectored vaccines encoding two blood-stage antigens: merozoite surface protein 1 (MSP1) and apical membrane antigen 1 (AMA1). Previously vaccinated volunteers show boosting of pre-existing antigen-specific memory B-cell (mBC) responses following CHMI. In contrast, unvaccinated malaria-naive control volunteers developed an mBC response against MSP1 but not AMA1. Serum IgG correlated with the mBC response after booster vaccination but this relationship was less well maintained following CHMI. A significant reduction in peripheral MSP1-specific mBC was observed at the point of diagnosis of blood-stage infection. This was coincident with a reduction in peripheral blood B-cell subsets expressing CXCR3 and elevated serum levels of interferon-γ and CXCL9, suggesting migration away from the periphery. These CHMI data confirm that mBC and antibody responses can be induced and boosted by blood-stage parasite exposure, in support of epidemiological studies on low-level parasite exposure.


Asunto(s)
Adenoviridae/genética , Antígenos de Protozoos/administración & dosificación , Linfocitos B/efectos de los fármacos , Inmunización , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/prevención & control , Proteínas de la Membrana/administración & dosificación , Proteína 1 de Superficie de Merozoito/administración & dosificación , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/administración & dosificación , Virus Vaccinia/genética , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/parasitología , Quimiocina CXCL9/sangre , Vectores Genéticos , Humanos , Esquemas de Inmunización , Inmunización Secundaria , Inmunoglobulina G/sangre , Memoria Inmunológica , Interferón gamma/sangre , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Falciparum/sangre , Malaria Falciparum/diagnóstico , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteína 1 de Superficie de Merozoito/genética , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Receptores CXCR3/sangre , Factores de Tiempo
4.
J Immunol ; 187(5): 2602-16, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21813775

RESUMEN

A central goal in vaccinology is the induction of high and sustained Ab responses. Protein-in-adjuvant formulations are commonly used to achieve such responses. However, their clinical development can be limited by the reactogenicity of some of the most potent preclinical adjuvants and the cost and complexity of licensing new adjuvants for human use. Also, few adjuvants induce strong cellular immunity, which is important for protection against many diseases, such as malaria. We compared classical adjuvants such as aluminum hydroxide to new preclinical adjuvants and adjuvants in clinical development, such as Abisco 100, CoVaccine HT, Montanide ISA720, and stable emulsion-glucopyranosyl lipid A, for their ability to induce high and sustained Ab responses and T cell responses. These adjuvants induced a broad range of Ab responses when used in a three-shot protein-in-adjuvant regimen using the model Ag OVA and leading blood-stage malaria vaccine candidate Ags. Surprisingly, this range of Ab immunogenicity was greatly reduced when a protein-in-adjuvant vaccine was used to boost Ab responses primed by a human adenovirus serotype 5 vaccine recombinant for the same Ag. This human adenovirus serotype 5-protein regimen also induced a more cytophilic Ab response and demonstrated improved efficacy of merozoite surface protein-1 protein vaccines against a Plasmodium yoelii blood-stage challenge. This indicates that the differential immunogenicity of protein vaccine adjuvants may be largely overcome by prior immunization with recombinant adenovirus, especially for adjuvants that are traditionally considered poorly immunogenic in the context of subunit vaccination and may circumvent the need for more potent chemical adjuvants.


Asunto(s)
Adenoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Vacunas contra la Malaria/inmunología , Vacunación/métodos , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunoglobulina G/análisis , Inmunoglobulina G/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
5.
J Immunol ; 185(12): 7583-95, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21098232

RESUMEN

Protein-in-adjuvant formulations and viral-vectored vaccines encoding blood-stage malaria Ags have shown efficacy in rodent malaria models and in vitro assays against Plasmodium falciparum. Abs and CD4(+) T cell responses are associated with protective efficacy against blood-stage malaria, whereas CD8(+) T cells against some classical blood-stage Ags can also have a protective effect against liver-stage parasites. No subunit vaccine strategy alone has generated demonstrable high-level efficacy against blood-stage infection in clinical trials. The induction of high-level Ab responses, as well as potent T and B cell effector and memory populations, is likely to be essential to achieve immediate and sustained protective efficacy in humans. This study describes in detail the immunogenicity of vaccines against P. falciparum apical membrane Ag 1 in rhesus macaques (Macaca mulatta), including the chimpanzee adenovirus 63 (AdCh63), the poxvirus modified vaccinia virus Ankara (MVA), and protein vaccines formulated in Alhydrogel or CoVaccine HT adjuvants. AdCh63-MVA heterologous prime-boost immunization induces strong and long-lasting multifunctional CD8(+) and CD4(+) T cell responses that exhibit a central memory-like phenotype. Three-shot (AdCh63-MVA-protein) or two-shot (AdCh63-protein) regimens induce memory B cells and high-titer functional IgG responses that inhibit the growth of two divergent strains of P. falciparum in vitro. Prior immunization with adenoviral vectors of alternative human or simian serotype does not affect the immunogenicity of the AdCh63 apical membrane Ag 1 vaccine. These data encourage the further clinical development and coadministration of protein and viral vector vaccine platforms in an attempt to induce broad cellular and humoral immune responses against blood-stage malaria Ags in humans.


Asunto(s)
Adenoviridae , Adyuvantes Inmunológicos , Antígenos de Protozoos/inmunología , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Poxviridae , Proteínas Protozoarias/inmunología , Virus Vaccinia , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Inmunoglobulina G/inmunología , Memoria Inmunológica/inmunología , Macaca mulatta , Malaria Falciparum/inmunología , Ratones , Vacunas de Subunidad/inmunología
6.
Front Immunol ; 10: 1254, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31214195

RESUMEN

The malaria genome encodes over 5,000 proteins and many of these have also been proposed to be potential vaccine candidates, although few of these have been tested clinically. RH5 is one of the leading blood-stage Plasmodium falciparum malaria vaccine antigens and Phase I/II clinical trials of vaccines containing this antigen are currently underway. Its likely mechanism of action is to elicit antibodies that can neutralize merozoites by blocking their invasion of red blood cells (RBC). However, many other antigens could also elicit neutralizing antibodies against the merozoite, and most of these have never been compared directly to RH5. The objective of this study was to compare a range of blood-stage antigens to RH5, to identify any antigens that outperform or synergize with anti-RH5 antibodies. We selected 55 gene products, covering 15 candidate antigens that have been described in the literature and 40 genes selected on the basis of bioinformatics functional prediction. We were able to make 20 protein-in-adjuvant vaccines from the original selection. Of these, S-antigen and CyRPA robustly elicited antibodies with neutralizing properties. Anti-CyRPA IgG generally showed additive GIA with anti-RH5 IgG, although high levels of anti-CyRPA-specific rabbit polyclonal IgG were required to achieve 50% GIA. Our data suggest that further vaccine antigen screening efforts are required to identify a second merozoite target with similar antibody-susceptibility to RH5.


Asunto(s)
Eritrocitos/inmunología , Eritrocitos/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Merozoítos/inmunología , Plasmodium falciparum/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Especificidad de Anticuerpos/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Femenino , Humanos , Inmunoglobulina G/inmunología , Malaria Falciparum/prevención & control , Ratones , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Polimorfismo Genético , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología
7.
Sci Rep ; 8(1): 3390, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29467399

RESUMEN

Heterologous prime-boost vaccination with viral vectors simian adenovirus 63 (ChAd63) and Modified Vaccinia Ankara (MVA) induces potent T cell and antibody responses in humans. The 8-week regimen demonstrates significant efficacy against malaria when expressing the pre-erythrocytic malaria antigen Thrombospondin-Related Adhesion Protein fused to a multiple epitope string (ME-TRAP). We tested these vaccines in 7 new 4- and 8- week interval schedules to evaluate safety and immunogenicity of multiple ChAd63 ME-TRAP priming vaccinations (denoted A), multiple MVA ME-TRAP boosts (denoted M) and alternating vectors. All regimens exhibited acceptable reactogenicity and CD8+ T cell immunogenicity was enhanced with a 4-week interval (AM) and with incorporation of additional ChAd63 ME-TRAP vaccination at 4- or 8-weeks (AAM or A_A_M). Induction of TRAP antibodies was comparable between schedules. T cell immunity against the ChAd63 hexon did not affect T cell responses to the vaccine insert, however pre-vaccination ChAd63-specific T cells correlated with reduced TRAP antibodies. Vaccine-induced antibodies against MVA did not affect TRAP antibody induction, and correlated positively with ME-TRAP-specific T cells. This study identifies potentially more effective immunisation regimens to assess in Phase IIa trials and demonstrates a degree of flexibility with the timing of vectored vaccine administration, aiding incorporation into existing vaccination programmes.


Asunto(s)
Epítopos/inmunología , Vectores Genéticos/inmunología , Hígado/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Adenovirus de los Simios/inmunología , Adolescente , Adulto , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Inmunización Secundaria/métodos , Masculino , Persona de Mediana Edad , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Vacunación/métodos , Vaccinia/inmunología , Virus Vaccinia/inmunología , Adulto Joven
8.
JCI Insight ; 2(12)2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28614791

RESUMEN

BACKGROUND: Plasmodium vivax is the most widespread human malaria geographically; however, no effective vaccine exists. Red blood cell invasion by the P. vivax merozoite depends on an interaction between the Duffy antigen receptor for chemokines (DARC) and region II of the parasite's Duffy-binding protein (PvDBP_RII). Naturally acquired binding-inhibitory antibodies against this interaction associate with clinical immunity, but it is unknown whether these responses can be induced by human vaccination. METHODS: Safety and immunogenicity of replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and modified vaccinia virus Ankara (MVA) viral vectored vaccines targeting PvDBP_RII (Salvador I strain) were assessed in an open-label dose-escalation phase Ia study in 24 healthy UK adults. Vaccines were delivered by the intramuscular route in a ChAd63-MVA heterologous prime-boost regimen using an 8-week interval. RESULTS: Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. PvDBP_RII-specific ex-vivo IFN-γ T cell, antibody-secreting cell, memory B cell, and serum IgG responses were observed after the MVA boost immunization. Vaccine-induced antibodies inhibited the binding of vaccine homologous and heterologous variants of recombinant PvDBP_RII to the DARC receptor, with median 50% binding-inhibition titers greater than 1:100. CONCLUSION: We have demonstrated for the first time to our knowledge that strain-transcending antibodies can be induced against the PvDBP_RII antigen by vaccination in humans. These vaccine candidates warrant further clinical evaluation of efficacy against the blood-stage P. vivax parasite. TRIAL REGISTRATION: Clinicaltrials.gov NCT01816113. FUNDING: Support was provided by the UK Medical Research Council, UK National Institute of Health Research Oxford Biomedical Research Centre, and the Wellcome Trust.

9.
Front Immunol ; 6: 348, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26217340

RESUMEN

Malaria vaccine development has largely focused on Plasmodium falciparum; however, a reawakening to the importance of Plasmodium vivax has spurred efforts to develop vaccines against this difficult to treat and at times severe form of relapsing malaria, which constitutes a significant proportion of human malaria cases worldwide. The almost complete dependence of P. vivax red blood cell invasion on the interaction of the P. vivax Duffy-binding protein region II (PvDBP_RII) with the human Duffy antigen receptor for chemokines (DARC) makes this antigen an attractive vaccine candidate against blood-stage P. vivax. Here, we generated both preclinical and clinically compatible adenoviral and poxviral vectored vaccine candidates expressing the Salvador I allele of PvDBP_RII - including human adenovirus serotype 5 (HAdV5), chimpanzee adenovirus serotype 63 (ChAd63), and modified vaccinia virus Ankara (MVA) vectors. We report on the antibody and T cell immunogenicity of these vaccines in mice or rabbits, either used alone in a viral vectored prime-boost regime or in "mixed-modality" adenovirus prime - protein-in--adjuvant boost regimes (using a recombinant PvDBP_RII protein antigen formulated in Montanide(®)ISA720 or Abisco(®)100 adjuvants). Antibodies induced by these regimes were found to bind to native parasite antigen from P. vivax infected Thai patients and were capable of inhibiting the binding of PvDBP_RII to its receptor DARC using an in vitro binding inhibition assay. In recent years, recombinant ChAd63 and MVA vectors have been quickly translated into human clinical trials for numerous antigens from P. falciparum as well as a growing number of other pathogens. The vectors reported here are immunogenic in small animals, elicit antibodies against PvDBP_RII, and have recently entered clinical trials, which will provide the first assessment of the safety and immunogenicity of the PvDBP_RII antigen in humans.

10.
J Leukoc Biol ; 95(2): 369-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24163420

RESUMEN

New tools are required to expedite the development of an effective vaccine against the blood-stage infection with the human malaria parasite Plasmodium falciparum. This work describes the assessment of the ADRB assay in a mouse model, characterizing the functional interaction between antimalarial serum antibodies and FcRs upon neutrophils. We describe a reproducible, antigen-specific assay, dependent on functional FcR signaling, and show that ADRB activity is induced equally by IgG1 and IgG2a isotypes and is modulated by blocking FcR function. However, following immunization of mice with the blood-stage vaccine candidate antigen MSP142, no measurable ADRB activity was induced against PEMS and neither was vaccine efficacy modulated against Plasmodium yoelii blood-stage challenge in γ(-/-) mice compared with WT mice. In contrast, following a primary, nonlethal P. yoelii parasite challenge, serum from vaccinated mice and nonimmunized controls showed anti-PEMS ADRB activity. Upon secondary challenge, nonimmunized γ(-/-) mice showed a reduced ability to control blood-stage parasitemia compared with immunized γ(-/-) mice; however, WT mice, depleted of their neutrophils, did not lose their ability to control infection. Thus, whereas neutrophil-induced ADRB against PEMS does not appear to play a role in protection against P. yoelii rodent malaria, induction of ADRB activity after challenge suggests that antigen targets of anti-PEMS ADRB activity remain to be established, as well as further supporting the observation that ADRB activity to P. falciparum arises following repeated natural exposure.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Inmunoensayo/métodos , Malaria/inmunología , Malaria/parasitología , Neutrófilos/inmunología , Plasmodium yoelii/inmunología , Estallido Respiratorio/inmunología , Animales , Antígenos de Protozoos/inmunología , Femenino , Humanos , Inmunoglobulina G/metabolismo , Ratones , Parásitos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal/inmunología , Resultado del Tratamiento
11.
PLoS One ; 9(9): e107903, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25254500

RESUMEN

The development of protective vaccines against many difficult infectious pathogens will necessitate the induction of effective antibody responses. Here we assess humoral immune responses against two antigens from the blood-stage merozoite of the Plasmodium falciparum human malaria parasite--MSP1 and AMA1. These antigens were delivered to healthy malaria-naïve adult volunteers in Phase Ia clinical trials using recombinant replication-deficient viral vectors--ChAd63 to prime the immune response and MVA to boost. In subsequent Phase IIa clinical trials, immunized volunteers underwent controlled human malaria infection (CHMI) with P. falciparum to assess vaccine efficacy, whereby all but one volunteer developed low-density blood-stage parasitemia. Here we assess serum antibody responses against both the MSP1 and AMA1 antigens following i) ChAd63-MVA immunization, ii) immunization and CHMI, and iii) primary malaria exposure in the context of CHMI in unimmunized control volunteers. Responses were also assessed in a cohort of naturally-immune Kenyan adults to provide comparison with those induced by a lifetime of natural malaria exposure. Serum antibody responses against MSP1 and AMA1 were characterized in terms of i) total IgG responses before and after CHMI, ii) responses to allelic variants of MSP1 and AMA1, iii) functional growth inhibitory activity (GIA), iv) IgG avidity, and v) isotype responses (IgG1-4, IgA and IgM). These data provide the first in-depth assessment of the quality of adenovirus-MVA vaccine-induced antibody responses in humans, along with assessment of how these responses are modulated by subsequent low-density parasite exposure. Notable differences were observed in qualitative aspects of the human antibody responses against these malaria antigens depending on the means of their induction and/or exposure of the host to the malaria parasite. Given the continued clinical development of viral vectored vaccines for malaria and a range of other diseases targets, these data should help to guide further immuno-monitoring studies of vaccine-induced human antibody responses.


Asunto(s)
Adenoviridae/inmunología , Antígenos de Protozoos/inmunología , Inmunidad Humoral/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Vacunación/métodos , Virus Vaccinia/inmunología , Adenoviridae/genética , Animales , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Sangre/parasitología , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Vacunas contra la Malaria/genética , Malaria Falciparum/sangre , Malaria Falciparum/inmunología , Pan troglodytes , Plasmodium falciparum/inmunología , Plasmodium falciparum/fisiología , Especificidad de la Especie , Virus Vaccinia/genética
12.
Expert Rev Vaccines ; 12(4): 365-78, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23560918

RESUMEN

Viral vectored vaccine delivery platforms have traditionally been used for the induction of cellular rather than humoral immunity. However, in recent years, recombinant adenoviral and poxviral vectored vaccines have been optimized to induce B-cell responses, resulting in the demonstration of high-titer antibody responses in a wide variety of animal species. These approaches have now been translated, confirming the induction of substantial levels of antigen-specific IgG in a series of Phase I human clinical trials targeting HIV-1 and Plasmodium falciparum malaria. To further improve the induction of antibodies, mixed-modality regimens based on recombinant viral and protein/adjuvant vaccines are now being assessed. However, limited data exist about the underlying mechanisms mediating the induction of B-cell responses by these subunit vaccines and their ability to influence the qualitative aspects of vaccine-induced B-cell populations and immunoglobulin. Future studies in this area are needed to guide the rational design of antibody-inducing subunit vaccine strategies.


Asunto(s)
Adenoviridae/genética , Portadores de Fármacos/administración & dosificación , Sistemas de Liberación de Medicamentos , Poxviridae/genética , Vacunación/métodos , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Ensayos Clínicos Fase I como Asunto , Infecciones por VIH/prevención & control , Humanos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control
13.
Sci Rep ; 3: 1706, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23609325

RESUMEN

Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.


Asunto(s)
Formación de Anticuerpos/inmunología , Antimaláricos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Merozoítos/inmunología , Plasmodium berghei/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Línea Celular , Femenino , Células HEK293 , Humanos , Inmunización/métodos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Conejos , Receptores de IgG/inmunología , Roedores/inmunología
14.
PLoS One ; 7(9): e44943, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22984589

RESUMEN

Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.


Asunto(s)
Adenoviridae/genética , Proteína de Unión al Complemento C4b/metabolismo , Malaria/prevención & control , Linfocitos T/citología , Adyuvantes Inmunológicos/genética , Animales , Antígenos de Protozoos/genética , Femenino , Vectores Genéticos , Vacunas contra la Malaria/genética , Proteína 1 de Superficie de Merozoito/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Estructura Terciaria de Proteína , Conejos , Receptores de IgG/metabolismo , Linfocitos T/metabolismo , Linfocitos T/virología , Vacunas/genética
15.
Clin Vaccine Immunol ; 17(7): 1066-73, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20484570

RESUMEN

Tuberculosis (TB) remains a threat to global health. While advances in diagnostics and treatment are crucial to the containment of the epidemic, it is likely that elimination of the disease can only be achieved through vaccination. Vaccine-induced protection from Mycobacterium tuberculosis is dependent, at least in part, on a robust Th1 response, yet little is known of the ability of TB vaccines to induce other T-cell subsets which may influence vaccine efficacy. Interleukin-17A (IL-17A) is a proinflammatory cytokine produced by Th17 cells which has been associated with both immune pathology and protection against infectious disease. Following vaccination with MVA85A, a viral vector vaccine aimed at enhancing immune responses to M. tuberculosis, antigen-specific IL-17A-producing T cells were induced in the peripheral blood of healthy volunteers. These T cells are detected later than gamma interferon (IFN-gamma)-secreting T cells and are of a low magnitude. Preexisting immune responses to mycobacterial antigens were associated with higher CD4(+) CD25(hi) CD39(+) T-cell levels in the periphery and a reduced capacity to produce IL-17A following immunization. These data highlight the intricate balance of effector and regulatory immune responses induced by vaccination and that preexisting immunity to mycobacterial antigens may affect the composition of vaccine-induced T-cell subsets.


Asunto(s)
Vacuna BCG/inmunología , Interleucina-17/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Vacunas contra la Tuberculosis/inmunología , Adolescente , Adulto , Ensayos Clínicos como Asunto , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Observación , Resultado del Tratamiento , Vacunación , Adulto Joven
16.
Vaccine ; 28(44): 7167-78, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20937436

RESUMEN

Subunit vaccination modalities tend to induce particular immune effector responses. Viral vectors are well known for their ability to induce strong T cell responses, while protein-adjuvant vaccines have been used primarily for induction of antibody responses. Here, we demonstrate in mice using a Plasmodium falciparum merozoite surface protein 1 (PfMSP1) antigen that novel regimes combining adenovirus and poxvirus vectored vaccines with protein antigen in Montanide ISA720 adjuvant can achieve simultaneous antibody and T cell responses which equal, or in some cases surpass, the best immune responses achieved by either the viral vectors or the protein vaccine alone. Such broad responses can be achieved either using three-stage vaccination protocols, or with an equally effective two-stage protocol in which viral vectors are admixed with protein and adjuvant, and were apparent despite the use of a protein antigen that represented only a portion of the viral vector antigen. We describe further possible advantages of viral vectors in achieving consistent antibody priming, enhanced antibody avidity, and cytophilic isotype skew. These data strengthen the evidence that tailored combinations of vaccine platforms can achieve desired combinations of immune responses, and further encourage the co-administration of antibody-inducing recombinant protein vaccines with T cell- and antibody-inducing recombinant viral vectors as one strategy that may achieve protective blood-stage malaria immunity in humans.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD8-positivos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Proteína 1 de Superficie de Merozoito/inmunología , Adenoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antiprotozoarios/inmunología , Afinidad de Anticuerpos , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Malaria Falciparum/inmunología , Manitol/análogos & derivados , Manitol/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ácidos Oléicos/farmacología , Plasmodium falciparum/inmunología , Poxviridae/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA