Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1130054, 2023.
Article in English | MEDLINE | ID: mdl-37056783

ABSTRACT

Introduction: A highly efficacious and durable vaccine against malaria is an essential tool for global malaria eradication. One of the promising strategies to develop such a vaccine is to induce robust CD8+ T cell mediated immunity against malaria liver-stage parasites. Methods: Here we describe a novel malaria vaccine platform based on a secreted form of the heat shock protein, gp96-immunoglobulin, (gp96-Ig) to induce malaria antigen specific, memory CD8+ T cells. Gp96-Ig acts as an adjuvant to activate antigen presenting cells (APCs) and chaperone peptides/antigens to APCs for cross presentation to CD8+ T cells. Results: Our study shows that vaccination of mice and rhesus monkeys with HEK-293 cells transfected with gp96-Ig and two well-known Plasmodium falciparum CSP and AMA1 (PfCA) vaccine candidate antigens, induces liver-infiltrating, antigen specific, memory CD8+ T cell responses. The majority of the intrahepatic CSP and AMA1 specific CD8+ T cells expressed CD69 and CXCR3, the hallmark of tissue resident memory T cells (Trm). Also, we found intrahepatic, antigen-specific memory CD8+ T cells secreting IL-2, which is relevant for maintenance of effective memory responses in the liver. Discussion: Our novel gp96-Ig malaria vaccine strategy represents a unique approach to induce liver-homing, antigen-specific CD8+ T cells critical for Plasmodium liver-stage protection.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Malaria , Humans , Heat-Shock Proteins/metabolism , HEK293 Cells , CD8-Positive T-Lymphocytes , Immunoglobulins/metabolism , Antigens, Protozoan , Malaria/prevention & control , Malaria/metabolism
2.
PLoS One ; 16(9): e0256980, 2021.
Article in English | MEDLINE | ID: mdl-34495988

ABSTRACT

BACKGROUND: A DNA-prime/human adenovirus serotype 5 (HuAd5) boost vaccine encoding Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) and Pf apical membrane antigen-1 (PfAMA1), elicited protection in 4/15 (27%) of subjects against controlled human malaria infection (CHMI) that was statistically associated with CD8+ T cell responses. Subjects with high level pre-existing immunity to HuAd5 were not protected, suggesting an adverse effect on vaccine efficacy (VE). We replaced HuAd5 with chimpanzee adenovirus 63 (ChAd63), and repeated the study, assessing both the two-antigen (CSP, AMA1 = CA) vaccine, and a novel three-antigen (CSP, AMA1, ME-TRAP = CAT) vaccine that included a third pre-erythrocytic stage antigen [malaria multiple epitopes (ME) fused to the Pf thrombospondin-related adhesive protein (TRAP)] to potentially enhance protection. METHODOLOGY: This was an open label, randomized Phase 1 trial, assessing safety, tolerability, and VE against CHMI in healthy, malaria naïve adults. Forty subjects (20 each group) were to receive three monthly CA or CAT DNA priming immunizations, followed by corresponding ChAd63 boost four months later. Four weeks after the boost, immunized subjects and 12 infectivity controls underwent CHMI by mosquito bite using the Pf3D7 strain. VE was assessed by determining the differences in time to parasitemia as detected by thick blood smears up to 28-days post CHMI and utilizing the log rank test, and by calculating the risk ratio of each treatment group and subtracting from 1, with significance calculated by the Cochran-Mantel-Haenszel method. RESULTS: In both groups, systemic adverse events (AEs) were significantly higher after the ChAd63 boost than DNA immunizations. Eleven of 12 infectivity controls developed parasitemia (mean 11.7 days). In the CA group, 15 of 16 (93.8%) immunized subjects developed parasitemia (mean 12.0 days). In the CAT group, 11 of 16 (63.8%) immunized subjects developed parasitemia (mean 13.0 days), indicating significant protection by log rank test compared to infectivity controls (p = 0.0406) and the CA group (p = 0.0229). VE (1 minus the risk ratio) in the CAT group was 25% compared to -2% in the CA group. The CA and CAT vaccines induced robust humoral (ELISA antibodies against CSP, AMA1 and TRAP, and IFA responses against sporozoites and Pf3D7 blood stages), and cellular responses (IFN-γ FluoroSpot responses to CSP, AMA1 and TRAP) that were not associated with protection. CONCLUSIONS: This study demonstrated that the ChAd63 CAT vaccine exhibited significant protective efficacy, and confirmed protection was afforded by adding a third antigen (T) to a two-antigen (CA) formulation to achieve increased VE. Although the ChAd63-CAT vaccine was associated with increased frequencies of systemic AEs compared to the CA vaccine and, historically, compared to the HuAd5 vectored malaria vaccine encoding CSP and AMA1, they were transient and associated with increased vector dosing.


Subject(s)
Adenovirus Vaccines/immunology , Adenoviruses, Simian/immunology , Antigens, Protozoan/immunology , DNA, Protozoan/immunology , DNA, Recombinant/immunology , Immunization, Secondary/methods , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Adenovirus Vaccines/administration & dosage , Adenovirus Vaccines/adverse effects , Adenoviruses, Simian/genetics , Adult , Antigens, Protozoan/genetics , CD8-Positive T-Lymphocytes/immunology , DNA, Protozoan/genetics , Epitopes/genetics , Epitopes/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Healthy Volunteers , Humans , Immunogenicity, Vaccine/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Membrane Proteins/genetics , Protozoan Proteins/genetics , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Young Adult
3.
PLoS One ; 15(5): e0232234, 2020.
Article in English | MEDLINE | ID: mdl-32407410

ABSTRACT

Only a small fraction of the antigens expressed by malaria parasites have been evaluated as vaccine candidates. A successful malaria subunit vaccine will likely require multiple antigenic targets to achieve broad protection with high protective efficacy. Here we describe protective efficacy of a novel antigen, Plasmodium yoelii (Py) E140 (PyE140), evaluated against P. yoelii challenge of mice. Vaccines targeting PyE140 reproducibly induced up to 100% sterile protection in both inbred and outbred murine challenge models. Although PyE140 immunization induced high frequency and multifunctional CD8+ T cell responses, as well as CD4+ T cell responses, protection was mediated by PyE140 antibodies acting against blood stage parasites. Protection in mice was long-lasting with up to 100% sterile protection at twelve weeks post-immunization and durable high titer anti-PyE140 antibodies. The E140 antigen is expressed in all Plasmodium species, is highly conserved in both P. falciparum lab-adapted strains and endemic circulating parasites, and is thus a promising lead vaccine candidate for future evaluation against human malaria parasite species.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Immunization , Malaria/prevention & control , Plasmodium yoelii/physiology , Animals , Antigens, Protozoan/genetics , Cross Reactions , Female , Gene Expression Regulation , Mice , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology
4.
Malar J ; 16(1): 263, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28673287

ABSTRACT

BACKGROUND: A DNA-human Ad5 (HuAd5) prime-boost malaria vaccine has been shown to protect volunteers against a controlled human malaria infection. The potency of this vaccine, however, appeared to be affected by the presence of pre-existing immunity against the HuAd5 vector. Since HuAd5 seroprevalence is very high in malaria-endemic areas of the world, HuAd5 may not be the most appropriate malaria vaccine vector. This report describes the evaluation of the seroprevalence, immunogenicity and efficacy of three newly identified gorilla adenoviruses, GC44, GC45 and GC46, as potential malaria vaccine vectors. RESULTS: The seroprevalence of GC44, GC45 and GC46 is very low, and the three vectors are not efficiently neutralized by human sera from Kenya and Ghana, two countries where malaria is endemic. In mice, a single administration of GC44, GC45 and GC46 vectors expressing a murine malaria gene, Plasmodium yoelii circumsporozoite protein (PyCSP), induced robust PyCSP-specific T cell and antibody responses that were at least as high as a comparable HuAd5-PyCSP vector. Efficacy studies in a murine malaria model indicated that a prime-boost regimen with DNA-PyCSP and GC-PyCSP vectors can protect mice against a malaria challenge. Moreover, these studies indicated that a DNA-GC46-PyCSP vaccine regimen was significantly more efficacious than a DNA-HuAd5-PyCSP regimen. CONCLUSION: These data suggest that these gorilla-based adenovectors have key performance characteristics for an effective malaria vaccine. The superior performance of GC46 over HuAd5 highlights its potential for clinical development.


Subject(s)
Adenoviruses, Simian , Genetic Vectors/standards , Malaria Vaccines/immunology , Malaria/prevention & control , Adenovirus Infections, Human/epidemiology , Adenovirus Infections, Human/virology , Adenoviruses, Simian/genetics , Adenoviruses, Simian/immunology , Animals , Antibodies, Viral/blood , Disease Models, Animal , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Ghana/epidemiology , Gorilla gorilla , Humans , Interferon-gamma/blood , Kenya/epidemiology , Malaria/epidemiology , Malaria Vaccines/standards , Mice , Mice, Inbred BALB C , Plasmids , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Seroepidemiologic Studies , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Transgenes/immunology , United States/epidemiology
5.
Vaccine ; 35(31): 3865-3874, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28596090

ABSTRACT

Malaria caused by Plasmodium falciparum continues to threaten millions of people living in the tropical parts of the world. A vaccine that confers sterile and life-long protection remains elusive despite more than 30years of effort and resources invested in solving this problem. Antibodies to a malaria vaccine candidate circumsporozoite protein (CSP) can block invasion and can protect humans against malaria. We have manufactured the Falciparum Malaria Protein-013 (FMP013) vaccine based on the nearly full-length P. falciparum CSP 3D7 strain sequence. We report here immunogenicity and challenge data on FMP013 antigen in C57BL/6 mice formulated with two novel adjuvants of the Army Liposome Formulation (ALF) series and a commercially available adjuvant Montanide ISA 720 (Montanide) as a control. ALF is a liposomal adjuvant containing a synthetic monophosphoryl lipid A (3D-PHAD®). In our study, FMP013 was adjuvanted with ALF alone, ALF containing aluminum hydroxide (ALFA) or ALF containing QS-21 (ALFQ). Adjuvants ALF and ALFA induced similar antibody titers and protection against transgenic parasite challenge that were comparable to Montanide. ALFQ was superior to the other three adjuvants as it induced higher antibody titers with improved boosting after the third immunization, higher serum IgG2c titers, and enhanced protection. FMP013+ALFQ also augmented the numbers of splenic germinal center-derived activated B-cells and antibody secreting cells compared to Montanide. Further, FMP013+ALFQ induced antigen-specific IFN-γ ELISPOT activity, CD4+ T-cells and a TH1-biased cytokine profile. These results demonstrate that soluble CSP can induce a potent and sterile protective immune response when formulated with the QS-21 containing adjuvant ALFQ. Comparative mouse immunogenicity data presented here were used as the progression criteria for an ongoing non-human primate study and a regulatory toxicology study in preparation for a controlled human malaria infection (CHMI) trial.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Lipid A/analogs & derivatives , Liposomes/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Saponins/administration & dosage , Animals , Antibodies, Protozoan/blood , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Female , Interferon-gamma/metabolism , Lipid A/administration & dosage , Malaria Vaccines/administration & dosage , Mice, Inbred C57BL
6.
PLoS One ; 10(8): e0136109, 2015.
Article in English | MEDLINE | ID: mdl-26292257

ABSTRACT

BACKGROUND: Nearly 100% protection against malaria infection can be achieved in humans by immunization with P. falciparum radiation-attenuated sporozoites (RAS). Although it is thought that protection is mediated by T cell and antibody responses, only a few of the many pre-erythrocytic (sporozoite and liver stage) antigens that are targeted by these responses have been identified. METHODOLOGY: Twenty seven P. falciparum pre-erythrocytic antigens were selected using bioinformatics analysis and expression databases and were expressed in a wheat germ cell-free protein expression system. Recombinant proteins were recognized by plasma from RAS-immunized subjects, and 21 induced detectable antibody responses in mice and rabbit and sera from these immunized animals were used to characterize these antigens. All 21 proteins localized to the sporozoite: five localized to the surface, seven localized to the micronemes, cytoplasm, endoplasmic reticulum or nucleus, two localized to the surface and cytoplasm, and seven remain undetermined. PBMC from RAS-immunized volunteers elicited positive ex vivo or cultured ELISpot responses against peptides from 20 of the 21 antigens. CONCLUSIONS: These T cell and antibody responses support our approach of using reagents from RAS-immunized subjects to screen potential vaccine antigens, and have led to the identification of a panel of novel P. falciparum antigens. These results provide evidence to further evaluate these antigens as vaccine candidates. TRIAL REGISTRATION: ClinicalTrials.gov NCT00870987 ClinicalTrials.gov NCT00392015.


Subject(s)
Antigens, Protozoan/immunology , Erythrocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Erythrocytes/parasitology , Humans , Immunization , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/parasitology , Malaria Vaccines/pharmacology , Malaria, Falciparum/blood , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Rabbits , Sporozoites/immunology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
7.
PLoS One ; 8(10): e78407, 2013.
Article in English | MEDLINE | ID: mdl-24205224

ABSTRACT

A key strategy to a successful vaccine against malaria is to identify and develop new adjuvants that can enhance T-cell responses and improve protective immunity. Upon co-administration with a rodent malaria vaccine in mice, 7DW8-5, a recently identified novel analog of α-galactosylceramide (α-GalCer), enhances the level of malaria-specific protective immune responses more strongly than the parent compound. In this study, we sought to determine whether 7DW8-5 could provide a similar potent adjuvant effect on a candidate human malaria vaccine in the more relevant non-human primate (NHP) model, prior to committing to clinical development. The candidate human malaria vaccine, AdPfCA (NMRC-M3V-Ad-PfCA), consists of two non-replicating recombinant adenoviral (Ad) vectors, one expressing the circumsporozoite protein (CSP) and another expressing the apical membrane antigen-1 (AMA1) of Plasmodium falciparum. In several phase 1 clinical trials, AdPfCA was well tolerated and demonstrated immunogenicity for both humoral and cell-mediated responses. In the study described herein, 25 rhesus macaques received prime and boost intramuscular (IM) immunizations of AdPfCA alone or with an ascending dose of 7DW8-5. Our results indicate that 7DW8-5 is safe and well-tolerated and provides a significant enhancement (up to 9-fold) in malaria-specific CD8+ T-cell responses after both priming and boosting phases, supporting further clinical development.


Subject(s)
Adenoviridae/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , CD8-Positive T-Lymphocytes/immunology , Glycolipids/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/drug effects , Genetic Vectors/immunology , Macaca mulatta/immunology , Malaria, Falciparum/drug therapy , Male , Membrane Proteins/immunology , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Primates/immunology , Protozoan Proteins/immunology
8.
Hum Vaccin Immunother ; 9(10): 2165-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23899517

ABSTRACT

BACKGROUND: In a prior study, a DNA prime / adenovirus boost vaccine (DNA/Ad) expressing P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1) (NMRC-M3V-D/Ad-PfCA Vaccine) induced 27% protection against controlled human malaria infection (CHMI). To investigate the contribution of DNA priming, we tested the efficacy of adenovirus vaccine alone (NMRC-M3V-Ad-PfCA ) in a Phase 1 clinical trial. METHODOLOGY/PRINCIPAL FINDINGS: The regimen was a single intramuscular injection with two non-replicating human serotype 5 adenovectors encoding CSP and AMA1, respectively. One x 10 (10) particle units of each construct were combined prior to administration. The regimen was safe and well-tolerated. Four weeks later, 18 study subjects received P. falciparum CHMI administered by mosquito bite. None were fully protected although one showed delayed onset of parasitemia. Antibody responses were low, with geometric mean CSP ELISA titer of 381 (range<50-1626) and AMA1 ELISA of 4.95 µg/mL (range 0.2-38). Summed ex vivo IFN-γ ELISpot responses to overlapping peptides were robust, with geometric mean spot forming cells/million peripheral blood mononuclear cells [sfc/m] for CSP of 273 (range 38-2550) and for AMA1 of 1303 (range 435-4594). CD4+ and CD8+ T cell IFN-γ responses to CSP were positive by flow cytometry in 25% and 56% of the research subjects, respectively, and to AMA1 in 94% and 100%, respectively. SIGNIFICANCE: In contrast to DNA/Ad, Ad alone did not protect against CHMI despite inducing broad, cell-mediated immunity, indicating that DNA priming is required for protection by the adenovirus-vectored vaccine. ClinicalTrials.gov Identifier: NCT00392015.


Subject(s)
Adenoviruses, Human/genetics , Antigens, Protozoan/immunology , Genetic Vectors , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Humans , Injections, Intramuscular , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria Vaccines/genetics , Male , Membrane Proteins/genetics , Middle Aged , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Young Adult
9.
PLoS One ; 8(2): e55571, 2013.
Article in English | MEDLINE | ID: mdl-23457473

ABSTRACT

BACKGROUND: Gene-based vaccination using prime/boost regimens protects animals and humans against malaria, inducing cell-mediated responses that in animal models target liver stage malaria parasites. We tested a DNA prime/adenovirus boost malaria vaccine in a Phase 1 clinical trial with controlled human malaria infection. METHODOLOGY/PRINCIPAL FINDINGS: The vaccine regimen was three monthly doses of two DNA plasmids (DNA) followed four months later by a single boost with two non-replicating human serotype 5 adenovirus vectors (Ad). The constructs encoded genes expressing P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). The regimen was safe and well-tolerated, with mostly mild adverse events that occurred at the site of injection. Only one AE (diarrhea), possibly related to immunization, was severe (Grade 3), preventing daily activities. Four weeks after the Ad boost, 15 study subjects were challenged with P. falciparum sporozoites by mosquito bite, and four (27%) were sterilely protected. Antibody responses by ELISA rose after Ad boost but were low (CSP geometric mean titer 210, range 44-817; AMA1 geometric mean micrograms/milliliter 11.9, range 1.5-102) and were not associated with protection. Ex vivo IFN-γ ELISpot responses after Ad boost were modest (CSP geometric mean spot forming cells/million peripheral blood mononuclear cells 86, range 13-408; AMA1 348, range 88-1270) and were highest in three protected subjects. ELISpot responses to AMA1 were significantly associated with protection (p = 0.019). Flow cytometry identified predominant IFN-γ mono-secreting CD8+ T cell responses in three protected subjects. No subjects with high pre-existing anti-Ad5 neutralizing antibodies were protected but the association was not statistically significant. SIGNIFICANCE: The DNA/Ad regimen provided the highest sterile immunity achieved against malaria following immunization with a gene-based subunit vaccine (27%). Protection was associated with cell-mediated immunity to AMA1, with CSP probably contributing. Substituting a low seroprevalence vector for Ad5 and supplementing CSP/AMA1 with additional antigens may improve protection. TRIAL REGISTRATION: ClinicalTrials.govNCT00870987.


Subject(s)
Adenoviruses, Human/genetics , Antigens, Protozoan/genetics , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Membrane Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Vaccines, DNA/therapeutic use , Adenoviruses, Human/immunology , Adolescent , Adult , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunity, Cellular , Interferon-gamma/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Membrane Proteins/immunology , Middle Aged , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Young Adult
10.
PLoS One ; 7(4): e33920, 2012.
Article in English | MEDLINE | ID: mdl-22496772

ABSTRACT

The development of an effective malaria vaccine is a high global health priority. Vaccine vectors based on adenovirus type 5 are capable of generating robust and protective T cell and antibody responses in animal models and are currently being evaluated in clinical trials for HIV and malaria. They appear to be more effective in terms of inducing antigen-specific immune responses as compared with non-Ad5 serotype vectors. However, the high prevalence of neutralizing antibodies to Ad5 in the human population, particularly in the developing world, has the potential to limit the effectiveness of Ad5-based vaccines. We have generated novel Ad5-based vectors that precisely replace the hexon hypervariable regions with those derived from Ad43, a subgroup D serotype with low prevalence of neutralizing antibody in humans. We have demonstrated that these hexon-modified adenovectors are not neutralized efficiently by Ad5 neutralizing antibodies in vitro using sera from mice, rabbits and human volunteers. We have also generated hexon-modified adenovectors that express a rodent malaria parasite antigen, PyCSP, and demonstrated that they are as immunogenic as an unmodified vector. Furthermore, in contrast to the unmodified vector, the hexon-modified adenovectors induced robust T cell responses in mice with high levels of Ad5 neutralizing antibody. We also show that the hexon-modified vector can be combined with unmodified Ad5 vector in prime-boost regimens to induce protective responses in mice. Our data establish that these hexon-modified vectors are highly immunogenic even in the presence of pre-existing anti-adenovirus antibodies. These hexon-modified adenovectors may have advantages in sub-Saharan Africa where there is a high prevalence of Ad5 neutralizing antibody in the population.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Antibodies, Neutralizing/immunology , Capsid Proteins/immunology , Genetic Vectors/immunology , Immunity, Cellular/immunology , Viral Vaccines/immunology , Animals , Capsid Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genetic Engineering , Genetic Vectors/genetics , Humans , Immunization , Mice , Mice, Inbred BALB C , Neutralization Tests , Rabbits , T-Lymphocytes/immunology , Transgenes/immunology , Viral Vaccines/administration & dosage
11.
PLoS One ; 6(10): e24586, 2011.
Article in English | MEDLINE | ID: mdl-22003383

ABSTRACT

BACKGROUND: Models of immunity to malaria indicate the importance of CD8+ T cell responses for targeting intrahepatic stages and antibodies for targeting sporozoite and blood stages. We designed a multistage adenovirus 5 (Ad5)-vectored Plasmodium falciparum malaria vaccine, aiming to induce both types of responses in humans, that was tested for safety and immunogenicity in a Phase 1 dose escalation trial in Ad5-seronegative volunteers. METHODOLOGY/PRINCIPAL FINDINGS: The NMRC-M3V-Ad-PfCA vaccine combines two adenovectors encoding circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). Group 1 (n = 6) healthy volunteers received one intramuscular injection of 2×10∧10 particle units (1×10∧10 each construct) and Group 2 (n = 6) a five-fold higher dose. Transient, mild to moderate adverse events were more pronounced with the higher dose. ELISpot responses to CSP and AMA1 peaked at 1 month, were higher in the low dose (geomean CSP = 422, AMA1 = 862 spot forming cells/million) than in the high dose (CSP = 154, p = 0.049, AMA1 = 423, p = 0.045) group and were still positive at 12 months in a number of volunteers. ELISpot depletion assays identified dependence on CD4+ or on both CD4+ and CD8+ T cells, with few responses dependent only on CD8+ T cells. Intracellular cytokine staining detected stronger CD8+ than CD4+ T cell IFN-γ responses (CSP p = 0.0001, AMA1 p = 0.003), but similar frequencies of multifunctional CD4+ and CD8+ T cells secreting two or more of IFN-γ, TNF-α or IL-2. Median fluorescence intensities were 7-10 fold higher in triple than single secreting cells. Antibody responses were low but trended higher in the high dose group and did not inhibit growth of cultured P. falciparum blood stage parasites. SIGNIFICANCE: As found in other trials, adenovectored vaccines appeared safe and well-tolerated at doses up to 1×10∧11 particle units. This is the first demonstration in humans of a malaria vaccine eliciting strong CD8+ T cell IFN-γ responses. TRIAL REGISTRATION: ClinicalTrials.govNCT00392015.


Subject(s)
Adenoviridae/genetics , Antigens, Protozoan/adverse effects , Antigens, Protozoan/immunology , Genetic Vectors/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Adolescent , Adult , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Immunologic , Female , Gene Expression , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Interferon-gamma/metabolism , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Male , Membrane Proteins/adverse effects , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Peptide Fragments/immunology , Protozoan Proteins/adverse effects , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Young Adult
12.
PLoS One ; 6(10): e25868, 2011.
Article in English | MEDLINE | ID: mdl-22003411

ABSTRACT

BACKGROUND: A protective malaria vaccine will likely need to elicit both cell-mediated and antibody responses. As adenovirus vaccine vectors induce both these responses in humans, a Phase 1/2a clinical trial was conducted to evaluate the efficacy of an adenovirus serotype 5-vectored malaria vaccine against sporozoite challenge. METHODOLOGY/PRINCIPAL FINDINGS: NMRC-MV-Ad-PfC is an adenovirus vector encoding the Plasmodium falciparum 3D7 circumsporozoite protein (CSP). It is one component of a two-component vaccine NMRC-M3V-Ad-PfCA consisting of one adenovector encoding CSP and one encoding apical membrane antigen-1 (AMA1) that was evaluated for safety and immunogenicity in an earlier study (see companion paper, Sedegah et al). Fourteen Ad5 seropositive or negative adults received two doses of NMRC-MV-Ad-PfC sixteen weeks apart, at 1 x 1010 particle units per dose. The vaccine was safe and well tolerated. All volunteers developed positive ELISpot responses by 28 days after the first immunization (geometric mean 272 spot forming cells/million[sfc/m]) that declined during the following 16 weeks and increased after the second dose to levels that in most cases were less than the initial peak (geometric mean 119 sfc/m). CD8+ predominated over CD4+ responses, as in the first clinical trial. Antibody responses were poor and like ELISpot responses increased after the second immunization but did not exceed the initial peak. Pre-existing neutralizing antibodies (NAb) to Ad5 did not affect the immunogenicity of the first dose, but the fold increase in NAb induced by the first dose was significantly associated with poorer antibody responses after the second dose, while ELISpot responses remained unaffected. When challenged by the bite of P. falciparum-infected mosquitoes, two of 11 volunteers showed a delay in the time to patency compared to infectivity controls, but no volunteers were sterilely protected. SIGNIFICANCE: The NMRC-MV-Ad-PfC vaccine expressing CSP was safe and well tolerated given as two doses, but did not provide sterile protection. TRIAL REGISTRATION: ClinicalTrials.gov NCT00392015.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/adverse effects , Protozoan Proteins/immunology , Adolescent , Adult , Antigens, Protozoan/adverse effects , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Dose-Response Relationship, Immunologic , Female , Gene Expression , Humans , Malaria Vaccines/genetics , Male , Membrane Proteins/adverse effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Plasmodium falciparum/cytology , Protozoan Proteins/genetics , Sporozoites/immunology , Young Adult
13.
Malar J ; 10: 65, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21410955

ABSTRACT

BACKGROUND: Despite years of effort, a licensed malaria vaccine is not yet available. One of the obstacles facing the development of a malaria vaccine is the extensive heterogeneity of many of the current malaria vaccine antigens. To counteract this antigenic diversity, an effective malaria vaccine may need to elicit an immune response against multiple malaria antigens, thereby limiting the negative impact of variability in any one antigen. Since most of the malaria vaccine antigens that have been evaluated in people have not elicited a protective immune response, there is a need to identify additional protective antigens. In this study, the efficacy of three pre-erythrocytic stage malaria antigens was evaluated in a Plasmodium yoelii/mouse protection model. METHODS: Mice were immunized with plasmid DNA and vaccinia virus vectors that expressed one, two or all three P. yoelii vaccine antigens. The immunized mice were challenged with 300 P. yoelii sporozoites and evaluated for subsequent infection. RESULTS: Vaccines that expressed any one of the three antigens did not protect a high percentage of mice against a P. yoelii challenge. However, vaccines that expressed all three antigens protected a higher percentage of mice than a vaccine that expressed PyCSP, the most efficacious malaria vaccine antigen. Dissection of the multi-antigen vaccine indicated that protection was primarily associated with two of the three P. yoelii antigens. The protection elicited by a vaccine expressing these two antigens exceeded the sum of the protection elicited by the single antigen vaccines, suggesting a potential synergistic interaction. CONCLUSIONS: This work identifies two promising malaria vaccine antigen candidates and suggests that a multi-antigen vaccine may be more efficacious than a single antigen vaccine.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium yoelii/immunology , Rodent Diseases/prevention & control , Vaccines, DNA/immunology , Animals , Antigens, Protozoan/genetics , Disease Models, Animal , Female , Humans , Malaria Vaccines/genetics , Mice , Plasmodium yoelii/genetics , Plasmodium yoelii/pathogenicity , Vaccines, DNA/genetics
14.
Vaccine ; 28(18): 3201-10, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20188680

ABSTRACT

An effective malaria vaccine remains a global health priority. Recombinant adenoviruses are a promising vaccine platform, and Plasmodium falciparum apical membrane antigen 1 (AMA1) and merozoite surface protein 1-42 (MSP1(42)) are leading blood stage vaccine candidates. We evaluated the importance of surface antigen localization and glycosylation on the immunogenicity of adenovector delivered AMA1 and MSP1(42) and assessed the ability of these vaccines to induce functional antibody responses capable of inhibiting parasite growth in vitro. Adenovector delivery induced unprecedented levels of biologically active antibodies in rabbits as indicated by the parasite growth inhibition assay. These responses were as potent as published results using any other vaccine system, including recombinant protein in adjuvant. The cell surface associated and glycosylated forms of AMA1 and MSP1(42) elicited 99% and 60% inhibition of parasite growth, respectively. Antigens that were expressed at the cell surface and glycosylated were much better than intracellular antigens at inducing antibody responses. Good T cell responses were observed for all forms of AMA1 and MSP1(42). Antigen-specific antibody responses, but typically not T cell responses, were boosted by a second administration of adenovector. These data highlight the importance of rational vaccine design and support the advancement of adenovector delivery technology for a malaria vaccine.


Subject(s)
Adenoviruses, Human/genetics , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Genetic Vectors , Malaria Vaccines/immunology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/growth & development , Protozoan Proteins/immunology , Animals , Antigens, Protozoan/genetics , Female , Glycosylation , Immunization, Secondary/methods , Malaria Vaccines/genetics , Membrane Proteins/genetics , Merozoite Surface Protein 1/genetics , Mice , Mice, Inbred BALB C , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Rabbits , T-Lymphocytes/immunology
15.
Vaccine ; 28(17): 3055-65, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-19879998

ABSTRACT

We previously reported the capacity of the cationic lipid-based formulation, Vaxfectin, to enhance the immunogenicity and protective efficacy of a low dose plasmid DNA vaccine against Plasmodium yoelii malaria in mice. Here, we have extended this finding to human Plasmodium falciparum genes, evaluating the immune enhancing effect of Vaxfectin formulation on a mixture, designated CSLAM, of five plasmid DNA vaccines encoding antigens from the sporozoite (PfCSP, PfSSP2/TRAP), intrahepatic (PfLSA1), and erythrocytic (PfAMA1, PfMSP1) life cycle stages of P. falciparum administered at 2, 10 or 50microg doses. Vaxfectin formulation enhanced both antibody and cellular immune responses to each component of the multi-antigen vaccine mixture, as assessed by ELISA, IFAT, and IFN-gamma ELIspot, respectively. There was no apparent antigenic competition, as indicated by comparison of responses induced in mice immunized with PfCSP vs. CSLAM. These data showing that Vaxfectin can enhance the immunogenicity of plasmid DNA vaccines administered at low doses per body weight, and in combinations, has important clinical implications for the development of a vaccine against malaria, as well as against other public health threats.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Protozoan/blood , Malaria Vaccines/immunology , Phosphatidylethanolamines/administration & dosage , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Female , Immunoassay/methods , Malaria Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Plasmodium falciparum/genetics , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Vaccines, DNA/administration & dosage
16.
Malar J ; 6: 135, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17925026

ABSTRACT

The present study has evaluated the immunogenicity of single or multiple Plasmodium falciparum (Pf) antigens administered in a DNA prime/poxvirus boost regimen with or without the poloxamer CRL1005 in rhesus monkeys. Animals were primed with PfCSP plasmid DNA or a mixture of PfCSP, PfSSP2/TRAP, PfLSA1, PfAMA1 and PfMSP1-42 (CSLAM) DNA vaccines in PBS or formulated with CRL1005, and subsequently boosted with ALVAC-Pf7, a canarypox virus expressing the CSLAM antigens. Cell-mediated immune responses were evaluated by IFN-gamma ELIspot and intracellular cytokine staining, using recombinant proteins and overlapping synthetic peptides. Antigen-specific and parasite-specific antibody responses were evaluated by ELISA and IFAT, respectively. Immune responses to all components of the multi-antigen mixture were demonstrated following immunization with either DNA/PBS or DNA/CRL1005, and no antigen interference was observed in animals receiving CSLAM as compared to PfCSP alone. These data support the down-selection of the CSLAM antigen combination. CRL1005 formulation had no apparent effect on vaccine-induced T cell or antibody responses, either before or after viral boost. In high responder monkeys, CD4+IL-2+ responses were more predominant than CD8+ T cell responses. Furthermore, CD8+ IFN-gamma responses were detected only in the presence of detectable CD4+ T cell responses. Overall, this study demonstrates the potential for multivalent Pf vaccines based on rational antigen selection and combination, and suggests that further formulation development to increase the immunogenicity of DNA encoded antigens is warranted.


Subject(s)
Antigens, Protozoan/immunology , Immunization, Secondary/methods , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Poxviridae/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/genetics , Immunization , Macaca mulatta , Malaria Vaccines/immunology , Plasmids , Vaccines, DNA/administration & dosage
17.
Vaccine ; 24(11): 1921-7, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16298024

ABSTRACT

We evaluated the capacity of the cationic lipid based formulation, Vaxfectin, to enhance the immunogenicity and protective efficacy of DNA-based vaccine regimens in the Plasmodium yoelii murine malaria model. We immunized Balb/c mice with varying doses (0.4-50 microg) of plasmid DNA (pDNA) encoding the P. yoelii circumsporozoite protein (PyCSP), either in a homologous DNA/DNA regimen (D-D) or a heterologous prime-boost DNA-poxvirus regimen (D-V). At the lowest pDNA doses, Vaxfectin substantially enhanced IFA titers, ELISPOT frequencies, and protective efficacy. Clinical trials of pDNA vaccines have often used low pDNA doses based on a per kilogram weight basis. Formulation of pDNA vaccines in Vaxfectin may improve their potency in human clinical trials.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Phosphatidylethanolamines/pharmacology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunization, Secondary , Lymphocytes/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Phosphatidylethanolamines/administration & dosage , Protozoan Proteins/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccinia virus/genetics
18.
Exp Hematol ; 33(6): 699-705, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15911094

ABSTRACT

The treatment of type I diabetes by islet cell transplantation, while promising, remains restricted due to the incomplete efficacy and toxicity associated with current immunosuppression, and by limited organ availability. Given reports suggesting bone marrow derived stem cell plasticity, we sought to determine whether such cells could give rise to pancreatic islet cells in vivo. In the context of autoimmune diabetes, we transplanted unfractionated bone marrow from beta-gal trangenic donor mice into NOD mice prior to, at, and two weeks beyond the onset of disease. Successful bone marrow engraftment before diabetes onset prevented disease in all mice and for 1 year after transplant. However, despite obtaining full hematopoietic engraftment in over 50 transplanted mice, only one mouse became insulin independent, and no beta-Gal positive islets were detected in any of the mice. To test whether tolerance to islets was achieved, we injected islets obtained from the same allogeneic donor strain as the hematopoietic cells into 4 transplant recipients, and 2 had a reversion of their diabetes. Thus allogeneic bone marrow transplantation prevents autoimmune diabetes and tolerizes the recipient to donor islet grants, even in diabetic animals, yet the capacity of bone marrow derived cells to differentiate into functional islet cells, at least without additional manipulation, is limited in our model.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Hematopoietic Stem Cell Transplantation , Islets of Langerhans/physiopathology , Regeneration , Animals , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
19.
Metabolism ; 53(4): 403-4, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15045683

ABSTRACT

Resistin is a recently described secretory protein produced in adipocytes that is thought to be involved in insulin resistance, diabetes, and inflammation. While resistin can be detected in mouse and human serum, very little is known about the regulation of serum resistin levels, especially in humans. To test whether resistin levels are affected by type 1 diabetes mellitus (T1DM), we measured serum resistin levels in samples from 5 healthy volunteers and 6 patients with T1DM pre- and 3 months post-islet transplantation using a human resistin enzyme immunoassay (EIA). Interestingly, serum resistin levels were significantly higher in T1DM patients before transplantation compared to normal controls, but decreased to normal levels after islet transplantation. Thus, our results suggest that human resistin may be involved in the pathophysiology of T1DM and thereby reveal a heretofore unappreciated aspect of human resistin biology.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/surgery , Hormones, Ectopic/blood , Islets of Langerhans Transplantation , Female , Humans , Monitoring, Intraoperative , Resistin
20.
Radiology ; 230(1): 163-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14695391

ABSTRACT

PURPOSE: To develop and assess a technique for induction of C peptide-negative diabetes in adult nonhuman primates in preparation for preclinical investigation of type 1 diabetes treatments. MATERIALS AND METHODS: First, temporary embolization of the hepatic and gastric arteries was performed in 14 adult nonhuman primates (six cynomolgus, five rhesus, and three pigtail macaques). After embolization was confirmed with angiography, streptozotocin was injected at a dose of 50-70 mg/kg into the celiac artery and branches supplying the pancreas. The macaques then were given intravenous injections of arginine and glucose, and blood levels of insulin and C peptide were measured with an enzyme-linked immunosorbent assay to determine whether diabetes had been induced. RESULTS: All but one of the macaques developed persistent long-term C peptide-negative diabetes after the streptozotocin injection. One macaque did not develop diabetes after the initial injection and was given a second dose of streoptozotocin, which did induce diabetes. None of the macaques showed any symptoms of hepatic or renal injury, and only one died (of gastric dilatation 5 days after the procedure). CONCLUSION: Streptozotocin injection after temporary embolization of the hepatic and gastric arteries is a safe and reproducible method for inducing C peptide-negative diabetes in adult nonhuman primates in preparation for preclinical investigation of type 1 diabetes treatments.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Animals , Embolization, Therapeutic , Injections, Intra-Arterial , Primates , Streptozocin/administration & dosage , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...