Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
PLoS One ; 6(9): e24413, 2011.
Article in English | MEDLINE | ID: mdl-21949716

ABSTRACT

BACKGROUND: In a previous Phase 1/2b malaria vaccine trial testing the 3D7 isoform of the malaria vaccine candidate Merozoite surface protein 2 (MSP2), parasite densities in children were reduced by 62%. However, breakthrough parasitemias were disproportionately of the alternate dimorphic form of MSP2, the FC27 genotype. We therefore undertook a dose-escalating, double-blinded, placebo-controlled Phase 1 trial in healthy, malaria-naïve adults of MSP2-C1, a vaccine containing recombinant forms of the two families of msp2 alleles, 3D7 and FC27 (EcMSP2-3D7 and EcMSP2-FC27), formulated in equal amounts with Montanide® ISA 720 as a water-in-oil emulsion. METHODOLOGY/PRINCIPAL FINDINGS: The trial was designed to include three dose cohorts (10, 40, and 80 µg), each with twelve subjects receiving the vaccine and three control subjects receiving Montanide® ISA 720 adjuvant emulsion alone, in a schedule of three doses at 12-week intervals. Due to unexpected local reactogenicity and concern regarding vaccine stability, the trial was terminated after the second immunisation of the cohort receiving the 40 µg dose; no subjects received the 80 µg dose. Immunization induced significant IgG responses to both isoforms of MSP2 in the 10 µg and 40 µg dose cohorts, with antibody levels by ELISA higher in the 40 µg cohort. Vaccine-induced antibodies recognised native protein by Western blots of parasite protein extracts and by immunofluorescence microscopy. Although the induced anti-MSP2 antibodies did not directly inhibit parasite growth in vitro, IgG from the majority of individuals tested caused significant antibody-dependent cellular inhibition (ADCI) of parasite growth. CONCLUSIONS/SIGNIFICANCE: As the majority of subjects vaccinated with MSP2-C1 developed an antibody responses to both forms of MSP2, and that these antibodies mediated ADCI provide further support for MSP2 as a malaria vaccine candidate. However, in view of the reactogenicity of this formulation, further clinical development of MSP2-C1 will require formulation of MSP2 in an alternative adjuvant. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry 12607000552482.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Life Cycle Stages/immunology , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Mannitol/analogs & derivatives , Oleic Acids/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Adolescent , Adult , Animals , Antibodies/immunology , Antigens, Protozoan/adverse effects , Chemistry, Pharmaceutical , Cohort Studies , Dose-Response Relationship, Immunologic , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Malaria Vaccines/adverse effects , Mannitol/chemistry , Plasmodium falciparum/growth & development , Protein Isoforms/adverse effects , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protozoan Proteins/adverse effects , Young Adult
2.
Vaccine ; 28(10): 2236-2242, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20051276

ABSTRACT

A Phase 1 trial was conducted in malaria-naïve adults to evaluate the recombinant protein vaccine apical membrane antigen 1-Combination 1 (AMA1-C1) formulated in Montanide ISA 720 (SEPPIC, France), a water-in-oil adjuvant. Vaccinations were halted early due to a formulation issue unrelated to stability or potency. Twenty-four subjects (12 in each group) were enrolled and received 5 or 20 microg protein at 0 and 3 months and four subjects were enrolled and received one vaccination of 80 microg protein. After first vaccination, nearly all subjects experienced mild to moderate local reactions and six experienced delayed local reactions occurring at Day 9 or later. After the second vaccination, three subjects experienced transient grade 3 (severe) local reactions; the remainder experienced grade 1 or 2 local reactions. All related systemic reactogenicity was grade 1 or 2, except one instance of grade 3 malaise. Anti-AMA1-C1 antibody responses were dose dependent and seen following each vaccination, with mean antibody levels 2-3 fold higher in the 20 microg group compared to the 5 microg group at most time points. In vitro growth-inhibitory activity was a function of the anti-AMA1 antibody titer. AMA1-C1 formulated in ISA 720 is immunogenic in malaria-naïve Australian adults. It is reasonably tolerated, though some transient, severe, and late local reactions are seen.


Subject(s)
Adjuvants, Immunologic/adverse effects , Antigens, Protozoan/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Mannitol/analogs & derivatives , Membrane Proteins/immunology , Oleic Acids/adverse effects , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Australia , Dose-Response Relationship, Immunologic , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Immunization, Secondary/methods , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Male , Mannitol/administration & dosage , Mannitol/adverse effects , Membrane Proteins/administration & dosage , Middle Aged , Oleic Acids/administration & dosage , Protozoan Proteins/administration & dosage , Young Adult
3.
Vaccine ; 26(52): 6864-73, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-18930094

ABSTRACT

Apical Membrane Antigen 1 (AMA1) and Merozoite Surface Protein 1 (MSP1) were produced as a recombinant fusion protein and formulated with the adjuvant Montanide ISA 720 with the aim of replicating the structure present in the parasite protein. A previous trial with this construct demonstrated the vaccine was safe and immunogenic but was associated with injection site reactogenicity. This Phase 1a dose-escalating, double blind, randomized, controlled trial of PfCP2.9/Montanide ISA 720 was conducted to evaluate alternative dose levels and vaccination schedules, with a pre-formulated vaccine that had undergone more in-depth and frequent quality control and stability analysis. The trial was conducted in seventy healthy Chinese malaria-naïve volunteers between January 2006 and January 2007. The objective was to assess the safety, reactogenicity and immunogenicity of 5, 20 and 50microg of PfCP2.9/ISA 720 under 2 different schedules. The most common adverse event was injection site tenderness (53%). The frequency and severity of adverse events was similar in both vaccination schedules. Antibody responses were induced and remained elevated throughout the study in volunteers receiving vaccine (p<0.001). Although high antibody titers as measured by ELISA to the PfCP2.9 immunogen were observed, biological function of these antibodies was not reflected by the in vitro inhibition of parasite growth, and there was limited recognition of fixed parasites in an immunofluorescence assay. At all three dose levels and both schedules, this formulation of PfCP2.9/ISA 720 is well tolerated, safe and immunogenic; however no functional activity against the parasite was observed.


Subject(s)
Calcium-Binding Proteins/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/immunology , Protein Kinases/immunology , Protozoan Proteins/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/pharmacology , Adolescent , Adult , Antibodies, Protozoan/analysis , Antibodies, Protozoan/biosynthesis , Chemistry, Pharmaceutical , Dose-Response Relationship, Immunologic , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Follow-Up Studies , Humans , Immunization Schedule , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/genetics , Male , Middle Aged , Mutant Chimeric Proteins/immunology , Pichia/chemistry , Pichia/immunology , Sample Size , Young Adult
4.
PLoS Clin Trials ; 2(4): e12, 2007.
Article in English | MEDLINE | ID: mdl-17415408

ABSTRACT

OBJECTIVES: To assess the safety and immunogenicity of two vaccines, MSP1(42)-FVO/Alhydrogel and MSP1(42)-3D7/Alhydrogel, targeting blood-stage Plasmodium falciparum parasites. DESIGN: A Phase 1 open-label, dose-escalating study. SETTING: Quintiles Phase 1 Services, Lenexa, Kansas between July 2004 and November 2005. PARTICIPANTS: Sixty healthy malaria-naïve volunteers 18-48 y of age. INTERVENTIONS: The C-terminal 42-kDa region of merozoite surface protein 1 (MSP1(42)) corresponding to the two allelic forms present in FVO and 3D7 P. falciparum lines were expressed in Escherichia coli, refolded, purified, and formulated on Alhydrogel (aluminum hydroxide). For each vaccine, volunteers in each of three dose cohorts (5, 20, and 80 microg) were vaccinated at 0, 28, and 180 d. Volunteers were followed for 1 y. OUTCOME MEASURES: The safety of MSP1(42)-FVO/Alhydrogel and MSP1(42)-3D7/Alhydrogel was assessed. The antibody response to each vaccine was measured by reactivity to homologous and heterologous MSP1(42), MSP1(19), and MSP1(33) recombinant proteins and recognition of FVO and 3D7 parasites. RESULTS: Anti-MSP1(42) antibodies were detected by ELISA in 20/27 (74%) and 22/27 (81%) volunteers receiving three vaccinations of MSP1(42)-FVO/Alhydrogel or MSP1(42)-3D7/Alhydrogel, respectively. Regardless of the vaccine, the antibodies were cross-reactive to both MSP1(42)-FVO and MSP1(42)-3D7 proteins. The majority of the antibody response targeted the C-terminal 19-kDa domain of MSP1(42), although low-level antibodies to the N-terminal 33-kDa domain of MSP1(42) were also detected. Immunofluorescence microscopy of sera from the volunteers demonstrated reactivity with both FVO and 3D7 P. falciparum schizonts and free merozoites. Minimal in vitro growth inhibition of FVO or 3D7 parasites by purified IgG from the sera of the vaccinees was observed. CONCLUSIONS: The MSP1(42)/Alhydrogel vaccines were safe and well tolerated but not sufficiently immunogenic to generate a biologic effect in vitro. Addition of immunostimulants to the Alhydrogel formulation to elicit higher vaccine-induced responses in humans may be required for an effective vaccine.

5.
Infect Immun ; 74(5): 2706-16, 2006 May.
Article in English | MEDLINE | ID: mdl-16622207

ABSTRACT

Heterologous prime-boost immunization with DNA and various recombinant poxviruses encoding malaria antigens is capable of inducing strong cell-mediated immune responses and partial protection in human sporozoite challenges. Here we report a series of trials assessing recombinant fowlpox virus and modified vaccinia virus Ankara encoding the Plasmodium falciparum circumsporozoite protein in various prime-boost combinations, doses, and application routes. For the first time, these vaccines were administered intramuscularly and at doses of up to 5 x 10(8) PFU. Vaccines containing this antigen proved safe and induced modest immune responses but showed no evidence of efficacy in a sporozoite challenge.


Subject(s)
Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccines, Synthetic/immunology , Adult , Animals , Antibodies, Protozoan/blood , Clinical Trials as Topic , Glycosylphosphatidylinositols/physiology , Humans , Immunization, Secondary , Interferon-gamma/biosynthesis , Malaria Vaccines/adverse effects , Parasitemia/prevention & control , Poxviridae/genetics , Vaccines, Synthetic/adverse effects , Vaccinia virus/genetics
6.
Vaccine ; 24(15): 2850-9, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16434127

ABSTRACT

Heterologous prime-boost immunisation with RTS,S/AS02A and the poxvirus MVA-CS was evaluated in 18 healthy malaria-naïve subjects in Oxford. Both priming with RTS,S and boosting MVA-CS, and the reverse, were found to be safe and well tolerated. T cell responses as measured by IFN-gamma ex vivo ELISPOT were induced, but the responses were low to moderate in both groups, with heterologous boosting yielding only small increments in T cell immunogenicity and no increased antibody response. Protection against 3D7 Plasmodium falciparum sporozoite challenge 4 weeks after the final vaccination was equal for both regimens at 33% (95% C.I. 4.3-77.7%), with one subject remaining fully protected on rechallenge at 5 months.


Subject(s)
Immunization, Secondary , Malaria Vaccines/immunology , Malaria/prevention & control , Vaccinia virus/immunology , Adolescent , Adult , Antibodies, Protozoan/blood , Female , Humans , Interferon-gamma/biosynthesis , Malaria/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Male , Middle Aged , T-Lymphocytes/immunology
7.
Infect Immun ; 73(6): 3587-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908388

ABSTRACT

Highly purified subunit vaccines require potent adjuvants in order to elicit optimal immune responses. In a previous phase I trial, an alum formulation of ICC-1132, a malaria vaccine candidate comprising hepatitis B core (HBc) virus-like particle containing Plasmodium falciparum circumsporozoite (CS) protein epitopes, was shown to elicit Plasmodium falciparum-specific antibody and cellular responses. The present study was designed as a single-blind, escalating-dose phase I trial to evaluate the safety and immunogenicity of single intramuscular doses of ICC-1132 formulated in the more potent water-in-oil adjuvant Montanide ISA 720 (ICC-1132/ISA 720). The vaccine was safe and well tolerated, with transient injection site pain as the most frequent complaint. All vaccinees that received either 20 mug or 50 mug of ICC-1132/ISA 720 developed antiimmunogen and anti-HBc antibodies. The majority of volunteers in these two groups developed sporozoite-specific antibodies, predominantly of opsonizing immunoglobulin G subtypes. Peak titers and persistence of parasite-specific antibody following a single injection of the ISA 720 formulated vaccine were comparable to those obtained following two to three immunizations with alum-adsorbed ICC-1132. Peripheral blood mononuclear cells of ICC-1132/ISA 720 vaccinees proliferated and released cytokines (interleukin 2 and gamma interferon) when stimulated with recombinant P. falciparum CS protein, and CS-specific CD4(+) T-cell lines were established from volunteers with high levels of antibodies to the repeat region. The promising results obtained with a single dose of ICC-1132 formulated in Montanide ISA 720 encourage further clinical development of this malaria vaccine candidate.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hepatitis B Core Antigens/administration & dosage , Malaria Vaccines/administration & dosage , Mannitol/analogs & derivatives , Mannitol/administration & dosage , Oleic Acids/administration & dosage , Plasmodium falciparum/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Antibody Specificity , CD4-Positive T-Lymphocytes/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/classification , Interferon-gamma/biosynthesis , Lymphocyte Activation , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Molecular Sequence Data , Single-Blind Method
8.
Vaccine ; 23(7): 857-64, 2005 Jan 04.
Article in English | MEDLINE | ID: mdl-15603885

ABSTRACT

ICC-1132, a recombinant virus-like particle comprising of a modified hepatitis B core protein with a B cell (NANP) and two T cell epitopes of Plasmodium falciparum circumsporozoite protein (CSP), was administered i.m. as a single 50 microg dose in Seppic ISA 720 to 11 volunteers. Local reactogenicity and systemic side effects were acceptable with the predominant finding being mild pain at the injection site. This regimen induced anti-NANP antibodies in 10/11 and modest T cell responses. There was no evidence of protection from experimental challenge with P. falciparum sporozoites. Other formulations and/or multi-dose regimens will be required to enhance the immunogenicity and efficacy of ICC-1132.


Subject(s)
Erythrocytes/immunology , Erythrocytes/parasitology , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adult , Amino Acid Sequence , Animals , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Male , Sporozoites/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...