Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
Rev Sci Instrum ; 95(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38912913

RÉSUMÉ

We describe a new experimental system for direct measurements of the absolute saturation vapor pressures of liquid or solid samples. The setup allows the isolation of the sample under steady conditions in an ultra-high vacuum chamber, where the measurement of the sample's vapor pressure as a function of its temperature can be performed in a range around room temperature and in a pressure range defined only by the applied absolute pressure sensor. We characterize the setup and illustrate its capability to measure saturation vapor pressures as well as enthalpies of evaporation around room temperature with explicit measurements on four liquid compounds (diethyl phthalate, 1-decanol, 1-heptanol, and 1-hexanol) for which accurate vapor pressures have previously been reported.

2.
JCI Insight ; 2(1): e89154, 2017 01 12.
Article de Anglais | MEDLINE | ID: mdl-28097230

RÉSUMÉ

BACKGROUND: A radiation-attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) malaria vaccine, PfSPZ Vaccine, protected 6 of 6 subjects (100%) against homologous Pf (same strain as in the vaccine) controlled human malaria infection (CHMI) 3 weeks after 5 doses administered intravenously. The next step was to assess protective efficacy against heterologous Pf (different from Pf in the vaccine), after fewer doses, and at 24 weeks. METHODS: The trial assessed tolerability, safety, immunogenicity, and protective efficacy of direct venous inoculation (DVI) of 3 or 5 doses of PfSPZ Vaccine in non-immune subjects. RESULTS: Three weeks after final immunization, 5 doses of 2.7 × 105 PfSPZ protected 12 of 13 recipients (92.3% [95% CI: 48.0, 99.8]) against homologous CHMI and 4 of 5 (80.0% [10.4, 99.5]) against heterologous CHMI; 3 doses of 4.5 × 105 PfSPZ protected 13 of 15 (86.7% [35.9, 98.3]) against homologous CHMI. Twenty-four weeks after final immunization, the 5-dose regimen protected 7 of 10 (70.0% [17.3, 93.3]) against homologous and 1 of 10 (10.0% [-35.8, 45.6]) against heterologous CHMI; the 3-dose regimen protected 8 of 14 (57.1% [21.5, 76.6]) against homologous CHMI. All 22 controls developed Pf parasitemia. PfSPZ Vaccine was well tolerated, safe, and easy to administer. No antibody or T cell responses correlated with protection. CONCLUSIONS: We have demonstrated for the first time to our knowledge that PfSPZ Vaccine can protect against a 3-week heterologous CHMI in a limited group of malaria-naive adult subjects. A 3-dose regimen protected against both 3-week and 24-week homologous CHMI (87% and 57%, respectively) in this population. These results provide a foundation for developing an optimized immunization regimen for preventing malaria. TRIAL REGISTRATION: ClinicalTrials.gov NCT02215707. FUNDING: Support was provided through the US Army Medical Research and Development Command, Military Infectious Diseases Research Program, and the Naval Medical Research Center's Advanced Medical Development Program.


Sujet(s)
Paludisme à Plasmodium falciparum/thérapie , Plasmodium falciparum/effets des médicaments et des substances chimiques , Sporozoïtes/effets des médicaments et des substances chimiques , Vaccins atténués/administration et posologie , Administration par voie intraveineuse , Adulte , Femelle , Humains , Paludisme à Plasmodium falciparum/prévention et contrôle , Mâle , Plasmodium falciparum/génétique , Sporozoïtes/génétique , Lymphocytes T/immunologie , Vaccins atténués/usage thérapeutique , Séquençage du génome entier/méthodes
3.
Vaccine ; 32(19): 2251-9, 2014 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-24126211

RÉSUMÉ

BACKGROUND: Novel influenza viruses continue to pose a potential pandemic threat worldwide. In recent years, plants have been used to produce recombinant proteins, including subunit vaccines. A subunit influenza vaccine, HAC1, based on recombinant hemagglutinin from the 2009 pandemic A/California/04/2009 (H1N1) strain of influenza virus, has been manufactured using a plant virus-based transient expression technology in Nicotiana benthamiana plants and demonstrated to be immunogenic and safe in pre-clinical studies (Shoji et al., 2011). METHODS: A first-in-human, Phase 1, single-center, randomized, placebo-controlled, single-blind, dose escalation study was conducted to investigate safety, reactogenicity and immunogenicity of an HAC1 formulation at three escalating dose levels (15 µg, 45 µg and 90 µg) with and without Alhydrogel(®), in healthy adults 18-50 years of age (inclusive). Eighty participants were randomized into six study vaccine groups, a saline placebo group and an approved monovalent H1N1 vaccine group. Recipients received two doses of vaccine or placebo (except for the monovalent H1N1 vaccine cohort, which received a single dose of vaccine, later followed by a dose of placebo). RESULTS: The experimental vaccine was safe and well tolerated, and comparable to placebo and the approved monovalent H1N1 vaccine. Pain and tenderness at the injection site were the only local solicited reactions reported following vaccinations. Nearly all adverse events were mild to moderate in severity. The HAC1 vaccine was also immunogenic, with the highest seroconversion rates, based on serum hemagglutination-inhibition and virus microneutralization antibody titers, in the 90 µg non-adjuvanted HAC1 vaccine group after the second vaccine dose (78% and 100%, respectively). CONCLUSIONS: This is the first study demonstrating the safety and immunogenicity of a plant-produced subunit H1N1 influenza vaccine in healthy adults. The results support further clinical investigation of the HAC1 vaccine as well as demonstrate the feasibility of the plant-based technology for vaccine antigen production.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza/immunologie , Vaccins antigrippaux/usage thérapeutique , Grippe humaine/prévention et contrôle , Adulte , Anticorps antiviraux/sang , Femelle , Humains , Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/effets indésirables , Vaccins antigrippaux/immunologie , Mâle , Adulte d'âge moyen , Protéines recombinantes/immunologie , Méthode en simple aveugle , Nicotiana , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/effets indésirables , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/usage thérapeutique , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/effets indésirables , Vaccins synthétiques/immunologie , Vaccins synthétiques/usage thérapeutique , Jeune adulte
4.
Vaccine ; 31(43): 4975-83, 2013 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-24029408

RÉSUMÉ

BACKGROUND: Immunization with genetically engineered, attenuated malaria parasites (GAP) that arrest during liver infection confers sterile protection in mouse malaria models. A first generation Plasmodium falciparum GAP (Pf p52(-)/p36(-) GAP) was previously generated by deletion of two pre-erythrocytic stage-expressed genes (P52 and P36) in the NF54 strain. METHODS: A first-in-human, proof-of-concept, safety and immunogenicity clinical trial in six human volunteers was conducted. Exposure consisted of delivery of Pf p52(-)/p36(-) GAP sporozoites via infected Anopheles mosquito bite with a five-bite/volunteer exposure followed by an approximately 200-bite exposure/volunteer one month later. RESULTS: The exposures were well tolerated with mild to moderate local and systemic reactions. All volunteers remained blood stage negative after low dose exposure. Five volunteers remained blood stage negative after high dose exposure. One volunteer developed peripheral parasitemia twelve days after high dose exposure. Together the findings indicate that Pf p52(-)/p36(-) GAP was severely but not completely attenuated. All six volunteers developed antibodies to CSP. Furthermore, IFN-γ responses to whole sporozoites and multiple antigens were elicited in 5 of 6 volunteers, with both CD4 and CD8 cell cytokine production detected. CONCLUSION: Severe attenuation and favorable immune responses following administration of a first generation Pf p52(-)/p36(-) GAP suggests that further development of live-attenuated strains using genetic engineering should be pursued.


Sujet(s)
Anopheles/parasitologie , Immunisation/méthodes , Vaccins contre le paludisme/immunologie , Paludisme/prévention et contrôle , Plasmodium falciparum/immunologie , Sporozoïtes/immunologie , Adolescent , Adulte , Animaux , Effets secondaires indésirables des médicaments/anatomopathologie , Femelle , Délétion de gène , Gènes de protozoaire , Volontaires sains , Humains , Immunisation/effets indésirables , Vaccins contre le paludisme/administration et posologie , Vaccins contre le paludisme/effets indésirables , Vaccins contre le paludisme/génétique , Mâle , Plasmodium falciparum/génétique , Plasmodium falciparum/pathogénicité , Vaccins atténués/administration et posologie , Vaccins atténués/effets indésirables , Vaccins atténués/génétique , Vaccins atténués/immunologie , Jeune adulte
5.
Vaccine ; 28(31): 5135-44, 2010 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-19737527

RÉSUMÉ

Plasmodium falciparum Liver Stage Antigen 1 (LSA-1) is a pre-erythrocytic stage antigen. Our LSA-1 vaccine candidate is a recombinant protein with full-length C- and N-terminal flanking domains and two of the 17 amino acid repeats from the central repeat region termed "LSA-NRC." We describe the first Phase I/II study of this recombinant LSA-NRC protein formulated with either the AS01 or AS02 adjuvant system. We conducted an open-label Phase I/II study. Thirty-six healthy malaria-naïve adults received one of four formulations by intra-deltoid injection on a 0 and 1 month schedule; low dose (LD) LSA-NRC/AS01:10microg LSA-NRC/0.5ml AS01 (n=5), high dose (HD) LSA-NRC/AS01: 50microg LSA-NRC/0.5ml AS01 (n=13); LD LSA-NRC/AS02: 10microg LSA-NRC/0.5ml AS02 (n=5) and HD LSA-NRC/AS02: 50microg LSA-NRC/0.5ml AS02 (n=13). Two weeks post-second immunization, the high dose vaccinees and 6 non-immunized infectivity controls underwent experimental malaria sporozoite challenge. The vaccines showed a reassuring safety profile but were moderately reactogenic. There were no serious adverse events. All subjects seroconverted after the first immunization. Following the second immunization, LSA-1-specific CD4+ T cells producing two cytokines (IL-2 and IFN-gamma) were found by intra-cellular staining in all subjects in the LD LSA-NRC/AS01B group and in 3 of 5 subjects in the LD LSA-NRC/AS02 group. In contrast, the HD LSA-NRC/AS01 and HD LSA-NRC/AS02 group subjects had fewer LSA-1-specific CD4+ T cells, and minimal to no IFN-gamma responses. There was no increase in LSA-1-specific CD8+ T cells found in any group. Per protocol, 22 high dose vaccinees, but no low dose vaccinees, underwent P. falciparum homologous malaria challenge (3D7 clone). All vaccinees became parasitemic and there was no delay in their pre-patent period versus controls (p=0.95). LSA-NRC/AS01 and LSA-NRC/AS02 elicited antigen-specific antibody and CD4+ T cell responses, but elicited no protective immunity. Although the optimal antigen dose of LSA-NRC may not have been selected for the challenge portion of the protocol, further vaccine development based upon LSA-1 should not be excluded and should include alternative vaccine platforms able to elicit additional effector mechanisms such as CD8+ T cells.


Sujet(s)
Antigènes de protozoaire/immunologie , Lymphocytes T CD4+/immunologie , Vaccins contre le paludisme/immunologie , Paludisme à Plasmodium falciparum/prévention et contrôle , Adjuvants immunologiques/pharmacologie , Adulte , Anticorps antiprotozoaires/sang , Production d'anticorps , Femelle , Humains , Immunité cellulaire , Immunité humorale , Calendrier vaccinal , Rappel de vaccin , Interféron gamma/immunologie , Interleukine-2/immunologie , Vaccins contre le paludisme/administration et posologie , Vaccins contre le paludisme/effets indésirables , Paludisme à Plasmodium falciparum/immunologie , Mâle , Parasitémie/immunologie , Plasmodium falciparum/immunologie , Protéines recombinantes/immunologie , Sporozoïtes/immunologie , Jeune adulte
6.
J Infect Dis ; 200(3): 337-46, 2009 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-19569965

RÉSUMÉ

BACKGROUND: To further increase the efficacy of malaria vaccine RTS,S/AS02A, we tested the RTS,S antigen formulated using the AS01B Adjuvant System (GlaxoSmithKline Biologicals). METHODS: In a double-blind, randomized trial, 102 healthy volunteers were evenly allocated to receive RTS,S/AS01B or RTS,S/AS02A vaccine at months 0, 1, and 2 of the study, followed by malaria challenge. Protected vaccine recipients were rechallenged 5 months later. RESULTS: RTS,S/AS01B and RTS,S/AS02A were well tolerated and were safe. The efficacy of RTS,S/AS01B and RTS,S/AS02A was 50% (95% confidence interval [CI], 32.9%-67.1%) and 32% (95% CI, 17.6%-47.6%), respectively. At the time of initial challenge, the RTS,S/AS01B group had greater circumsporozoite protein (CSP)-specific immune responses, including higher immunoglobulin (Ig) G titers, higher numbers of CSP-specific CD4(+) T cells expressing 2 activation markers (interleukin-2, interferon [IFN]-gamma, tumor necrosis factor-alpha, or CD40L), and more ex vivo IFN-gamma enzyme-linked immunospots (ELISPOTs) than did the RTS,S/AS02A group. Protected vaccine recipients had a higher CSP-specific IgG titer (geometric mean titer, 188 vs 73 mug/mL; P < .001), higher numbers of CSP-specific CD4(+) T cells per 10(6) CD4(+) T cells (median, 963 vs 308 CSP-specific CD4(+) T cells/10(6) CD4(+) T cells; P < .001), and higher numbers of ex vivo IFN-gamma ELISPOTs (mean, 212 vs 96 spots/million cells; P < .001). At rechallenge, 4 of 9 vaccine recipients in each group were still completely protected. CONCLUSIONS: The RTS,S/AS01B malaria vaccine warrants comparative field trials with RTS,S/AS02A to determine the best formulation for the protection of children and infants. The association between complete protection and immune responses is a potential tool for further optimization of protection. Trial registration. ClinicalTrials.gov identifier NCT00075049.


Sujet(s)
Vaccins contre le paludisme , Paludisme à Plasmodium falciparum/prévention et contrôle , Plasmodium falciparum/immunologie , Adulte , Animaux , Anticorps antiprotozoaires/sang , Études transversales , Méthode en double aveugle , Études de suivi , Humains , Vaccins contre le paludisme/effets indésirables , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/normes , Paludisme à Plasmodium falciparum/immunologie , Mozambique/épidémiologie , Parasitémie , Facteurs temps
7.
PLoS One ; 4(4): e5254, 2009.
Article de Anglais | MEDLINE | ID: mdl-19390585

RÉSUMÉ

BACKGROUND: This Phase 1/2a study evaluated the safety, immunogenicity, and efficacy of an experimental malaria vaccine comprised of the recombinant Plasmodium falciparum protein apical membrane antigen-1 (AMA-1) representing the 3D7 allele formulated with either the AS01B or AS02A Adjuvant Systems. METHODOLOGY/PRINCIPAL FINDINGS: After a preliminary safety evaluation of low dose AMA-1/AS01B (10 microg/0.5 mL) in 5 adults, 30 malaria-naïve adults were randomly allocated to receive full dose (50 microg/0.5 mL) of AMA-1/AS01B (n = 15) or AMA-1/AS02A (n = 15), followed by a malaria challenge. All vaccinations were administered intramuscularly on a 0-, 1-, 2-month schedule. All volunteers experienced transient injection site erythema, swelling and pain. Two weeks post-third vaccination, anti-AMA-1 Geometric Mean Antibody Concentrations (GMCs) with 95% Confidence Intervals (CIs) were high: low dose AMA-1/AS01B 196 microg/mL (103-371 microg/mL), full dose AMA-1/AS01B 279 microg/mL (210-369 microg/mL) and full dose AMA-1/AS02A 216 microg/mL (169-276 microg/mL) with no significant difference among the 3 groups. The three vaccine formulations elicited equivalent functional antibody responses, as measured by growth inhibition assay (GIA), against homologous but not against heterologous (FVO) parasites as well as demonstrable interferon-gamma (IFN-gamma) responses. To assess efficacy, volunteers were challenged with P. falciparum-infected mosquitoes, and all became parasitemic, with no significant difference in the prepatent period by either light microscopy or quantitative polymerase chain reaction (qPCR). However, a small but significant reduction of parasitemia in the AMA-1/AS02A group was seen with a statistical model employing qPCR measurements. SIGNIFICANCE: All three vaccine formulations were found to be safe and highly immunogenic. These immune responses did not translate into significant vaccine efficacy in malaria-naïve adults employing a primary sporozoite challenge model, but encouragingly, estimation of parasite growth rates from qPCR data may suggest a partial biological effect of the vaccine. Further evaluation of the immunogenicity and efficacy of the AMA-1/AS02A formulation is ongoing in a malaria-experienced pediatric population in Mali. TRIAL REGISTRATION: www.clinicaltrials.gov NCT00385047.


Sujet(s)
Adjuvants immunologiques/administration et posologie , Antigènes de protozoaire/immunologie , Lipide A/analogues et dérivés , Vaccins contre le paludisme/administration et posologie , Paludisme à Plasmodium falciparum/prévention et contrôle , Protéines membranaires/immunologie , Protéines de protozoaire/immunologie , Saponines/administration et posologie , Adjuvants immunologiques/pharmacologie , Adolescent , Adulte , Allèles , Animaux , Antigènes de protozoaire/administration et posologie , Antigènes de protozoaire/génétique , Méthode en double aveugle , Association médicamenteuse , Test ELISA , Humains , Lipide A/administration et posologie , Lipide A/pharmacologie , Vaccins contre le paludisme/effets indésirables , Vaccins contre le paludisme/immunologie , Protéines membranaires/administration et posologie , Protéines membranaires/génétique , Adulte d'âge moyen , Plasmodium falciparum/immunologie , Plasmodium falciparum/métabolisme , Protéines de protozoaire/administration et posologie , Protéines de protozoaire/génétique , Saponines/pharmacologie
8.
Vaccine ; 26(18): 2191-202, 2008 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-18387719

RÉSUMÉ

BACKGROUND: Immunization with RTS,S/AS02 consistently protects some vaccinees against malaria infection in experimental challenges and in field trials. A brief immunization schedule against falciparum malaria would be compatible with the Expanded Programme on Immunization, or in combination with other prevention measures, interrupt epidemic malaria or protect individuals upon sudden travel to an endemic area. METHODS: We conducted an open label, Phase 2a trial of two different full dose schedules of RTS,S/AS02 in 40 healthy malaria-naïve adults. Cohort 1 (n=20) was immunized on a 0, 1, and 3 month schedule and Cohort 2 (n=20) on a 0, 7, and 28 day schedule. Three weeks later, 38 vaccinees and 12 unimmunized infectivity controls underwent malaria challenge. RESULTS: Both regimens had a good safety and tolerability profile. Peak GMCs of antibody to the circumsporozoite protein (CSP) were similar in Cohort 1 (78 microg/mL; 95% CI: 45-134) and Cohort 2 (65 microg/mL; 95% CI: 40-104). Vaccine efficacy for Cohort 1 was 45% (95% CI: 18-62%) and for Cohort 2, 39% (95% CI: 11-56%). Protected volunteers had a higher GMC of anti-CSP antibody (114 microg/mL) than did volunteers with a 2-day delay (70 microg/mL) or no delay (30 microg/mL) in the time to onset of parasitemia (Kruskal-Wallis, p=0.019). A trend was seen for higher CSP-specific IFN-gamma responses in PBMC from protected volunteers only in Cohort 1, but not in Cohort 2, for ex vivo and for cultured ELISPOT assays. CONCLUSION: In malaria-naïve adults, the efficacy of three-dose RTS,S/AS02 regimens on either a 0, 1, and 3 month schedule or an abbreviated 0, 7, and 28 day schedule was not discernibly different from two previously reported trials of two-dose regimens given at 0, 1 month that conferred 47% (95% CI: -19 to 76%) protection and in another trial 42% (95% CI: 5-63%). A strong association of CSP-specific antibody with protection against malaria challenge is observed and confirms similar observations made in other studies. Subsequent trials of adjuvanted RTS,S in African children and infants on a 0, 1, and 2 month schedule have demonstrated a favorable safety and efficacy profile.


Sujet(s)
Calendrier vaccinal , Vaccins contre le paludisme/immunologie , Paludisme à Plasmodium falciparum/prévention et contrôle , Adolescent , Adulte , Anticorps antiprotozoaires/sang , Cellules cultivées , Femelle , Humains , Interféron gamma/biosynthèse , Agranulocytes/immunologie , Vaccins contre le paludisme/administration et posologie , Vaccins contre le paludisme/effets indésirables , Mâle , Adulte d'âge moyen , Parasitémie/prévention et contrôle , Protéines de protozoaire/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...