Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 32575, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27624667

ABSTRACT

Malaria eradication will require a combination of vector control, chemotherapy and an easily administered vaccine. Sterile immunity can be elicited in humans by immunization with sporozoites, the infective stage injected by bite of the mosquito vector, however, whole parasite vaccines present formidable logistical challenges for production, storage and administration. The "gold standard" for infectious disease eradiation, the Smallpox Eradication Programme, utilized mass immunization using the skin scarification (SS) route. SS may more closely mimic the natural route of malaria infection initiated by sporozoites injected by mosquito bite which elicits both neutralizing antibodies and protective cell mediated immunity. We investigated the potential of SS immunization using a malaria repeat peptide containing a protective B cell epitope of Plasmodium falciparum, the most lethal human species, and delivery vehicles containing TLR agonists as adjuvants. In a murine model, SS immunization with peptide in combination with TLR-7/8 and -9 agonists elicited high levels of systemic sporozoite neutralizing antibody, Th1- type CD4+ T cells and resistance to challenge by bites of infected mosquitoes. SS provides the potential to elicit humoral immunity to target Plasmodium at multiple stages of its complex life cycle.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Sporozoites/drug effects , Adjuvants, Immunologic/administration & dosage , Animals , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Immunization , Life Cycle Stages/drug effects , Life Cycle Stages/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/immunology , Sporozoites/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
2.
Malar J ; 15: 201, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068454

ABSTRACT

BACKGROUND: Plasmodium circumsporozoite protein (CSP) is a major surface antigen present in the sporozoite (Spz) stage of a malaria parasite. RTS, S vaccine, the most clinically advanced malaria vaccine, consists of a large portion of Plasmodium falciparum CSP (PfCSP). A highly infectious, recombinant rodent malaria, Plasmodium yoelii parasite bearing a full-length PfCSP, PfCSP/Py Spz, was needed as a tool to evaluate the role of PfCSP in mediating, protective, anti-malaria immunity in a mouse model. METHODS: A transgenic parasite, PfCSP/Py Spz, was generated by inserting a construct expressing the PfCSP at the locus of the P. yoelii CSP gene by double cross-over homologous recombination. Then the biological and protective properties of PfCSP/Py Spz were determined. RESULTS: This PfCSP/Py parasite produced up to 30,000 Spz in mosquito salivary glands, which is equal or even higher than the number of Spz produced by wild-type P. yoelii parasites. Five bites of PfCSP/Py-infected mosquitoes could induce blood infection in BALB/c mice. CONCLUSIONS: The current study has demonstrated a successful establishment of a transgenic P. yoelii parasite clone that is able to express a full-length PfCSP, PfCSP/Py parasite. Importantly, this PfCSP/Py parasite can be as infectious as the wild-type P. yoelii parasite both in mosquito vector and in mouse, a mammalian host. A new transgenic parasite that expresses a full-length PfCSP may become a useful tool for researchers to investigate immunity against PfCSP in a mouse model.


Subject(s)
Culicidae/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/immunology , Plasmodium falciparum/genetics , Salivary Glands/parasitology , T-Lymphocytes/parasitology , Vaccines, Synthetic/immunology
3.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410104

ABSTRACT

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Subject(s)
Disease Models, Animal , Immunity, Humoral/immunology , Malaria, Falciparum/immunology , Mice, Transgenic/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Heterografts , Histocompatibility Antigens Class II , Humans , Malaria Vaccines , Mice , Protozoan Proteins/immunology
4.
Biomaterials ; 33(27): 6476-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22695068

ABSTRACT

Biomaterials that modulate innate and adaptive immune responses are receiving increasing interest as adjuvants for eliciting protective immunity against a variety of diseases. Previous results have indicated that self-assembling ß-sheet peptides, when fused with short peptide epitopes, can act as effective adjuvants and elicit robust and long-lived antibody responses. Here we investigated the mechanism of immunogenicity and the quality of antibody responses raised by a peptide epitope from Plasmodium falciparum circumsporozoite (CS) protein, (NANP)(3),conjugated to the self-assembling peptide domain Q11. The mechanism of adjuvant action was investigated in knockout mice with impaired MyD88, NALP3, TLR-2, or TLR-5 function, and the quality of antibodies raised against (NANP)(3)-Q11 was assessed using a transgenic sporozoite neutralizing (TSN) assay for malaria infection. (NANP)(3)-Q11 self-assembled into nanofibers, and antibody responses lasted up to 40 weeks in C57BL/6 mice. The antibody responses were T cell- and MyD88-dependent. Sera from mice primed with either irradiated sporozoites or a synthetic peptide, (T1BT*)(4)-P3C, and boosted with (NANP)(3)-Q11 showed significant increases in antibody titers and significant inhibition of sporozoite infection in TSN assays. In addition, two different epitopes could be self-assembled together without compromising the strength or duration of the antibody responses raised against either of them, making these materials promising platforms for self-adjuvanting multi-antigenic immunotherapies.


Subject(s)
Antibody Formation/immunology , Epitopes/immunology , Malaria/immunology , Nanofibers/chemistry , Peptides/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myeloid Differentiation Factor 88/metabolism , Nanofibers/ultrastructure , Oligopeptides/chemistry , Oligopeptides/immunology , Peptides/chemistry , Protein Structure, Secondary , Sporozoites/immunology , T-Lymphocytes/immunology
5.
Infect Immun ; 76(3): 1200-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160479

ABSTRACT

The irradiated-sporozoite vaccine elicits sterile immunity against Plasmodium parasites in experimental rodent hosts and human volunteers. Based on rodent malaria models, it has been proposed that CD8+ T cells are the key protective effector mechanism required in sporozoite-induced immunity. To investigate the role of class II-restricted immunity in protective immunity, we immunized beta2-microglobulin knockout (beta2M-/-) mice with irradiated Plasmodium yoelii or P. berghei sporozoites. Sterile immunity was obtained in the CD8+-T-cell-deficient mice immunized with either P. berghei or P. yoelii sporozoites. beta2M-/- mice with the BALB/c (H-2d) genetic background as well as those with the C57BL (H-2b) genetic background were protected. Effector mechanisms included CD4+ T cells, mediated in part through the production of gamma interferon, and neutralizing antibodies that targeted the extracellular sporozoites. We conclude that in the absence of class I-restricted CD8+ T cells, sporozoite-induced protective immunity can be effectively mediated by class II-restricted immune effector mechanisms. These results support efforts to develop subunit vaccines that effectively elicit high levels of antibody and CD4+ T cells to target Plasmodium pre-erythrocytic stages.


Subject(s)
Malaria/prevention & control , Plasmodium berghei/immunology , Plasmodium yoelii/immunology , Sporozoites/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Liver/parasitology , Lymphocyte Depletion , Malaria/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Plasmodium berghei/radiation effects , Plasmodium yoelii/radiation effects , beta 2-Microglobulin/deficiency
6.
Infect Immun ; 74(12): 6929-39, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17030584

ABSTRACT

An effective malaria vaccine is needed to address the public health tragedy resulting from the high levels of morbidity and mortality caused by Plasmodium parasites. The first protective immune mechanism identified in the irradiated sporozoite vaccine, the "gold standard" for malaria preerythrocytic vaccines, was sporozoite-neutralizing antibody specific for the repeat region of the surface circumsporozoite (CS) protein. Previous phase I studies demonstrated that a branched peptide containing minimal T- and B-cell epitopes of Plasmodium falciparum CS protein elicited antirepeat antibody and CD4(+)-T-cell responses comparable to those observed in volunteers immunized with irradiated P. falciparum sporozoites. The current study compares the immunogenicity of linear versus tetrabranched peptides containing the same minimal T- and B-cell epitopes, T1BT*, comprised of a CS-derived universal Th epitope (T*) synthesized in tandem with the T1 and B repeats of P. falciparum CS protein. A simple 48-mer linear synthetic peptide was found to elicit antisporozoite antibody and gamma interferon-secreting T-cell responses comparable to the more complex tetrabranched peptides in inbred strains of mice. The linear peptide was also immunogenic in outbred nonhuman primates (Aotus nancymaae), eliciting antibody titers equivalent to those induced by tetrabranched peptides. Importantly, the 48-mer linear peptide administered in adjuvants suitable for human use elicited antibody-mediated protection against challenge with rodent malaria transgenic sporozoites expressing P. falciparum CS repeats. These findings support further evaluation of linear peptides as economical, safe, and readily produced malaria vaccines for the one-third of the world's population at risk of malaria infection.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Peptides/immunology , Plasmodium falciparum/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Aotidae , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Humans , Immunoglobulin G/blood , Malaria, Falciparum/immunology , Mice , Molecular Sequence Data , Sporozoites/immunology
7.
J Immunol ; 175(11): 7575-85, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301667

ABSTRACT

Peptide vaccines containing minimal epitopes of protective Ags provide the advantages of low cost, safety, and stability while focusing host responses on relevant targets of protective immunity. However, the limited complexity of malaria peptide vaccines raises questions regarding their equivalence to immune responses elicited by the irradiated sporozoite vaccine, the "gold standard" for protective immunity. A panel of CD4+ T cell clones was derived from volunteers immunized with a peptide vaccine containing minimal T and B cell epitopes of the Plasmodium falciparum circumsporozoite protein to compare these with previously defined CD4+ T cell clones from volunteers immunized with irradiated P. falciparum sporozoites. As found following sporozoite immunization, the majority of clones from the peptide-immunized volunteers recognized the T* epitope, a predicted universal T cell epitope, in the context of multiple HLA DR and DQ molecules. Peptide-induced T cell clones were of the Th0 subset, secreting high levels of IFN-gamma as well as variable levels of Th2-type cytokines (IL-4, IL-6). The T* epitope overlaps a polymorphic region of the circumsporozoite protein and strain cross-reactivity of the peptide-induced clones correlated with recognition of core epitopes overlapping the conserved regions of the T* epitope. Importantly, as found following sporozoite immunization, long-lived CD4+ memory cells specific for the T* epitope were detectable 10 mo after peptide immunization. These studies demonstrate that malaria peptides containing minimal epitopes can elicit human CD4+ T cells with fine specificity and potential effector function comparable to those elicited by attenuated P. falciparum sporozoites.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccines, Subunit/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , CD4-Positive T-Lymphocytes/microbiology , Clone Cells , Cytokines/biosynthesis , Cytokines/immunology , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunodominant Epitopes/immunology , Molecular Sequence Data , Vaccines, Synthetic/immunology
8.
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
9.
Infect Immun ; 72(11): 6519-27, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501783

ABSTRACT

We report the first phase I trial to assess the safety and immunogenicity of a malaria vaccine candidate, ICC-1132 (Malarivax), composed of a modified hepatitis B virus core protein (HBc) containing minimal epitopes of the Plasmodium falciparum circumsporozoite (CS) protein. When expressed in Escherichia coli, the recombinant ICC-1132 protein forms virus-like particles that were found to be highly immunogenic in preclinical studies of mice and monkeys. Twenty healthy adult volunteers received a 20- or a 50-microg dose of alum-adsorbed ICC-1132 administered intramuscularly at 0, 2, and 6 months. The majority of volunteers in the group receiving the 50-microg dose developed antibodies to CS repeats as well as to HBc. Malaria-specific T cells that secreted gamma interferon were also detected after a single immunization with ICC-1132-alum. These studies support ICC-1132 as a promising malaria vaccine candidate for further clinical testing using more-potent adjuvant formulations and confirm the potential of modified HBc virus-like particles as a delivery platform for vaccines against other human pathogens.


Subject(s)
Hepatitis B Core Antigens/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Alum Compounds , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Double-Blind Method , Epitopes , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B/metabolism , Hepatitis B Core Antigens/metabolism , Humans , Interferon-gamma/metabolism , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Middle Aged , Molecular Sequence Data , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Virion/genetics , Virion/immunology , Virion/metabolism
10.
Vaccine ; 21(3-4): 269-80, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12450702

ABSTRACT

We tested the clinical reactions to a synthetic, Plasmodium falciparum, circumsporozoite multiple antigen peptide (MAP) vaccine in 39 volunteers immunized two to three times over 2-8 months using a dose escalation design. Immediate pain at the injection site was associated with the adjuvant QS-21 (P<0.001), and delayed local inflammatory reactions were associated with high-titered circulating IgG anti-MAP antibody (P=0.03). Because two volunteers developed acute, systemic urticaria after the third immunization associated with development of serum IgE MAP antibody, we employed immediate-type hypersensitivity skin tests (ITH-STs) using intradermal injections of diluted MAP vaccine to identify persons sensitized to the vaccine. ITH-STs were negative in seven volunteers tested 27 days after the first vaccination, but six of these individuals developed positive wheal and flare reactions when tested 14 or 83 days after the second vaccination; IgE MAP antibody was detected in only one of them. Another cohort of 16 volunteers, including the 2 allergic individuals, were ITH-ST negative when first tested late after their second or third vaccination at 6-7 months. Five of five non-immunized persons were also ITH-ST negative. ITH-STs may help identify individuals sensitized to malaria peptides and at potential risk of developing systemic allergic reactions after re-vaccination.


Subject(s)
Antigens, Protozoan/immunology , Hypersensitivity, Delayed/chemically induced , Malaria Vaccines/adverse effects , Plasmodium falciparum/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Animals , Antigens, Protozoan/biosynthesis , Cohort Studies , Female , Human Experimentation , Humans , Hypersensitivity/etiology , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Intradermal Tests , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Male , Pain/chemically induced , Pain/immunology , Plasmodium falciparum/growth & development , Urticaria/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
11.
Vaccine ; 20(13-14): 1853-61, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11906775

ABSTRACT

During the testing of the safety and immunogenicity of an adjuvanted, synthetic Plasmodium falciparum CS multiple antigen peptide (MAP) vaccine, we investigated the potential for using cutaneous delayed-type hypersensitivity (DTH) reactions as a correlate of immune response. We evaluated 27 of our volunteers for DTH reactions to intradermal inoculation (0.02 ml) of several concentrations of the MAP vaccine and adjuvant control solutions. Induration was measured 2 days after skin tests were applied. Nine of 14 vaccinees (64%) with serum, high-titered anti-MAP antibody developed positive DTH (>or=5mm induration), that first appeared by 29 days after immunization and persisted for at least 3-6 months after 1-2 more immunizations. In contrast, DTH responses were negative in eight of eight vaccinees with no or low antibody titers, and in five of five non-immunized volunteers. Biopsies of positive DTH skin test sites were histologically compatible with a DTH reaction. We conclude that the presence of T cell functional activity reflected by a positive DTH skin test response to the MAP antigen serves as another marker for vaccine immunogenicity.


Subject(s)
Malaria Vaccines/pharmacology , Plasmodium falciparum/immunology , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Humans , Hypersensitivity, Delayed , Immunoglobulin G/biosynthesis , Intradermal Tests , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Middle Aged , Plasmodium falciparum/growth & development , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...